WO2008116308A1 - Quinolones et tétrahydroquinoléines et composés apparentés ayant une activité inhibitrice de nos - Google Patents

Quinolones et tétrahydroquinoléines et composés apparentés ayant une activité inhibitrice de nos Download PDF

Info

Publication number
WO2008116308A1
WO2008116308A1 PCT/CA2008/000569 CA2008000569W WO2008116308A1 WO 2008116308 A1 WO2008116308 A1 WO 2008116308A1 CA 2008000569 W CA2008000569 W CA 2008000569W WO 2008116308 A1 WO2008116308 A1 WO 2008116308A1
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
compound
mmol
mammal
comprises administering
Prior art date
Application number
PCT/CA2008/000569
Other languages
English (en)
Inventor
Shawn Maddaford
Jailall Ramnauth
Suman Rakhit
Joanne Patman
Subhash C. Annedi
John Andrews
Peter Dove
Sarah Silverman
Paul Renton
Original Assignee
Neuraxon, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=39775375&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2008116308(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Neuraxon, Inc. filed Critical Neuraxon, Inc.
Priority to JP2010500037A priority Critical patent/JP2010521527A/ja
Priority to MX2009010193A priority patent/MX2009010193A/es
Priority to CA002681771A priority patent/CA2681771A1/fr
Priority to CN200880017025A priority patent/CN101679397A/zh
Priority to EP08748081A priority patent/EP2139886A4/fr
Priority to AU2008232269A priority patent/AU2008232269A1/en
Priority to NZ580618A priority patent/NZ580618A/en
Publication of WO2008116308A1 publication Critical patent/WO2008116308A1/fr
Priority to IL201142A priority patent/IL201142A/en
Priority to ZA2009/07458A priority patent/ZA200907458B/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/06Antimigraine agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • A61P25/36Opioid-abuse
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings

Definitions

  • the field of the present invention relates to quinolones, tetrahydroquinolines, and related compounds and to their medical use.
  • Nitric oxide has diverse roles both in normal and pathological processes, including the regulation of blood pressure, in neurotransmission, and in the macrophage defense systems (Snyder et al., Scientific American, May 1992:68). NO is synthesized by three isoforms of nitric oxide synthase, a constitutive form in endothelial cells (eNOS), a constitutive form in neuronal cells (nNOS), and an inducible form found in macrophage cells (iNOS). These enzymes are homodimeric proteins that catalyze a five-electron oxidation of L-arginine, yielding NO and citrulline. The role of NO produced by each of the NOS isoforms is quite unique.
  • nNOS and iNOS Overstimulation or overproduction of individual NOS isoforms especially nNOS and iNOS, plays a role in several disorders, including septic shock, arthritis, diabetes, ischemia-reperfusion injury, pain, and various neurodegenerative diseases (Kerwin, et al., J. Med. Chem. 38:4343, 1995), while eNOS inhibition leads to unwanted effects such as enhanced white cell and platelet activation, hypertension and increased atherogenesis (Valance and Leiper, Nature Rev. Drug Disc. 2002, 1, 939).
  • NOS inhibitors have the potential to be used as therapeutic agents in many disorders.
  • the preservation of physiologically important nitric oxide synthase function suggests the desirability of the development of isoform- selective inhibitors that preferentially inhibit nNOS over eNOS.
  • Q is (CHR 6 ) 1-3 ;
  • R 1 and each R 6 is, independently, H, optionally substituted C 1-6 alkyl, optionally substituted C 1-4 alkaryl, optionally substituted C 1-4 alkheterocyclyl, or optionally substituted C 2-9 heterocyclyl; each of R 2 and R 3 is, independently, H, Hal, optionally substituted Cj -6 alkyl, optionally substituted C 6-10 aryl, optionally substituted C 1-6 alkaryl, optionally substituted C 2 9 heterocyclyl, hydroxy, optionally substituted C 1-6 alkoxy, optionally substituted C 1-6 thioalkoxy, (CH 2 ) r2 NHC(NH)R 2A , or (CH 2 ) r2 NHC(S)NHR 2A , or optionally substituted C 1-4 alkheterocyclyl, wherein r2 is an integer from 0 to 2, R 2A is optionally substituted C 1-6 alkyl, optionally substituted C 6-10 aryl, optionally substituted Ci -4 al
  • R 2 , R 3 , R 4 , and R 5 is (CH 2 ) r2 NHC(NH)R 2A or (CH 2 ) 24 NHC(S)NHR 2A ; or a pharmaceutically acceptable salt or prodrug thereof.
  • Q is (CHR 6 ) 1-3 ;
  • R 1 and each R 6 is, independently, H, optionally substituted Ci -6 alkyl, optionally substituted C 1-4 alkaryl, optionally substituted Ci -4 alkheterocyclyl, or optionally substituted C 2-9 heterocyclyl;
  • each of R 2 and R 3 is, independently, H, Hal, optionally substituted Ci -6 alkyl, optionally substituted C 6- io aryl, optionally substituted Ci -6 alkaryl, optionally substituted C 2-9 heterocyclyl, or optionally substituted Ci -4 alkheterocyclyl;
  • R 2 , R 3 , R 4 or R 5 has the formula:
  • each of X 1 , X 2 , X 4 , and X 5 is independently selected from O, S, NR 7 , N, or CR 8 ;
  • X 33 iiss sseelleecctteedd ffrroomm NN oorr CCRR 88 ;;
  • R 7 is H or optionally substituted Ci -6 alkyl
  • R 8 is H, Hal, optionally substituted Ci 6 alkyl, hydroxy, optionally substituted C 1-6 alkoxy, or optionally substituted Ci -6 thioalkoxy, wherein at least one of X 1 , X 2 , X 4 , and X 5 is not CR 8 .
  • R 2A may also have the formula:
  • each of X 1 and X 2 is independently selected from O, S, NH, N, or CH; wherein at least one of X 1 and X 2 is not CH.
  • X 1 is CH
  • X 2 is S
  • exemplary compounds are shown in Table 2.
  • the invention also features a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt or prodrug thereof, and a pharmaceutically acceptable excipient.
  • the invention further features a method of treating or preventing a condition in a mammal caused by the action of nitric oxide synthase (NOS), wherein said method comprises administering an effective amount of a compound of the invention or a pharmaceutically acceptable salt or prodrug thereof to said mammal, e.g., a human.
  • NOS nitric oxide synthase
  • Exemplary conditions include migraine headache (with or without aura), chronic tension type headache (CTTH), migraine with allodynia, medication overuse headache, neuropathic pain, AIDS associated painful neuropathy, chronic headache, central post-stroke pain (CPSP), medication-induced hyperalgesia or allodynia, acute pain, chronic pain, diabetic neuropathy, trigeminal neuralgia, chemotherapy induced neuropathic pain, bone cancer pain, chemical dependencies or addictions, CNS disorders, neurodegenerative diseases or nerve injury, cardiovascular related conditions, diabetic nephropathy, inflammatory diseases, or gastrointestinal disorders. Specific examples of these conditions are described herein. Preferred conditions are neuropathic pain, CTTH, visceral pain, and IBS.
  • the method may further include administering to the mammal an opioid, an antidepressant, an antiepileptic, a non-steroidal anti-inflammatory drug (NSAID), an antiarrhythmic, a GABA-B antagonist, an alpha-2-adrenergic receptor agonist, a serotonin 5HT 1B/1D agonist, an N-methyl-D-aspartate antagonist, a cholecystokinin B antagonist, a substance P antagonist, an anti-inflammatory compound, a DHP- sensitive L-type calcium channel antagonist, omega-conotoxin-sensitive N-type calcium channel antagonist, a P/Q-type calcium channel antagonist, an adenosine kinase antagonist, an adenosine receptor A 1 agonist, an adenosine receptor A 2a antagonist, an adenosine receptor A 3 agonist, an adenosine deaminase inhibitor, an adenosine nucleoside transport inhibito, a vanilloid VRl receptor
  • a compound of the invention selectively inhibits neuronal nitric oxide synthase (nNOS), particularly over endothelial nitric oxide synthase (eNOS) or inducible nitric oxide synthase (iNOS) or both.
  • nNOS neuronal nitric oxide synthase
  • eNOS endothelial nitric oxide synthase
  • iNOS inducible nitric oxide synthase
  • the IC 50 or K; value observed for the compound is at least 2 times lower for nNOS than for eNOS and/or iNOS. More preferably, the IC 50 or K 1 value is at least 5, 20, 50, or 100 times lower. In one embodiment, the IC 50 or K; value is between 2 times and 100 times lower. In another embodiment, the IC 50 or K, in eNOS is greater than 10 ⁇ M. More preferably eNOS IC 50 is greater than 20 ⁇ M, most preferably eNOS IC 50 or Ki is greater than 30 ⁇ M, as a threshold level of eNOS may be needed to avoid any direct eNOS mediated constriction of human vascular tissue. Exemplary compounds are described herein.
  • the invention also features a pharmaceutical composition that includes a compound of the invention and a pharmaceutically acceptable excipient.
  • the invention further features a method of treating or preventing a condition in a mammal, such as, for example, a human, caused by the action of nitric oxide synthase (NOS), e.g., nNOS, that includes administering an effective amount of a compound of the invention to the mammal.
  • NOS nitric oxide synthase
  • migraine headache with or without aura
  • CTH chronic tension type headache
  • CPSP central post-stroke pain
  • medication-induced hyperalgesia and/or allodynia e.g., opioid-induced hyperalgesia or triptan (5-HT1D/1B agonists)-induced hyperalgesia/allodynia, acute pain, chronic pain, diabetic neuropathy, trigeminal neuralgia, chemotherapy induced neuropathic pain (e.g., Paclitaxol, cis-Platin, Doxorubicin etc.), bone cancer pain, chemical dependencies or addictions (e.g., drug addiction, cocaine addiction, nicotine addiction, metamphetamine-induced neurotoxicity, ethanol tolerance, dependence, or withdrawal, or morphine/opioid induced tolerance, dependence, hyperalgesia, or withdrawal), CNS disorders (e.g., epilepsy
  • CNS disorders e.g., epilepsy
  • a compound of the invention can also be used in combination with one or more other therapeutic agents for the prevention or treatment of one of the aforementioned conditions.
  • agents useful in combination with a compound of the invention include opioids, antidepressants, antiepileptics, non-steroidal anti-inflammatory drugs (NSAIDs), antiarrhythmics, GABA-B antagonists, alpha-2-adrenergic receptor agonists, serotonin 5HT 1B/ID agonists, N-methyl-D-aspartate antagonists, cholecystokinin B antagonists, substance P antagonists (NKl), anti- inflammatory compounds, DHP-sensitive L-type calcium channel antagonists, omega-conotoxin- sensitive N-type calcium channel antagonists, P/Q-type calcium channel antagonists, adenosine kinase antagonists, adenosine receptor A 1 agonists, adenosine receptor A 2a antagonists, adenosine receptor A 3 agonists, adenosine deaminase inhibitors, adenosine nucleoside transport inhibitors, vanilloid VRl receptor agonists, can be opioid
  • therapeutic agents that are useful in combination with a compound of the invention are listed in Table 1.
  • Other classes include CB1/CB2 agonists, e.g., dexanabinol (HU-211), fatty acid amide hydrolase inhibitors, P2X purinergic blockers, and NGF antagonists.
  • NMDA antagonists in combination with nNOS inhibitors may be particularly useful in treating conditions such as inflammatory and neuropathic pain, traumatic brain injury and Parkinson's Disease (see Drug Discovery Today 2002: 7(7) 403-406).
  • Asymmetric or chiral centers may exist in any of the compounds of the present invention.
  • the present invention contemplates the various stereoisomers and mixtures thereof.
  • Individual stereoisomers of compounds of the present invention are prepared synthetically from commercially available starting materials which contain asymmetric or chiral centers or by preparation of mixtures of enantiomeric compounds followed by resolution well-known to those of ordinary skill in the art. These methods of resolution are exemplified by (1) attachment of a racemic mixture of enantiomers, designated (+/-), to a chiral auxiliary, separation of the resulting diastereomers by recrystallization or chromatography and liberation of the optically pure product from the auxiliary or (2) direct separation of the mixture of optical enantiomers on chiral chromatographic columns.
  • chiral compounds can be prepared by an asymmetric synthesis that favours the preparation of one enatiomer over the other.
  • a chiral pool synthesis (starting with an enantiomerically pure building block) can be used wherein the chiral group or center is retained in the intermediate or final product.
  • Enantiomers are designated herein by the symbols "R,” or “S,” depending on the configuration of substituents around the chiral atom.
  • enantiomers are designated as (+) or (-) depending on whether a solution of the enantiomer rotates the plane of polarized light clockwise or counterclockwise, respectively.
  • Geometric isomers may also exist in the compounds of the present invention.
  • the present invention contemplates the various geometric isomers and mixtures thereof resulting from the arrangement of substituents around a carbon-carbon double bond and designates such isomers as of the Z or E configuration, where the term "Z” represents substituents on the same side of the carbon-carbon double bond and the term “E” represents substituents on opposite sides of the carbon-carbon double bond. It is also recognized that for structures in which tautomeric forms are possible, the description of one tautomeric form is equivalent to the description of both, unless otherwise specified.
  • substituents and substitution patterns on the compounds of the invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art, as well as those methods set forth below, from readily available starting materials. If a substituent is itself substituted with more than one group, it is understood that these multiple groups may be on the same carbon or on different carbons, so long as a stable structure results.
  • acyl or "alkanoyl,” as used interchangeably herein, represent an alkyl group, as defined herein, or hydrogen attached to the parent molecular group through a carbonyl group, as defined herein, and is exemplified by formyl, acetyl, propionyl, butanoyl and the like.
  • exemplary unsubstituted acyl groups include from 2 to 7 carbons.
  • C x _ y alkaryl or "C x-y alkylenearyl,” as used herein, represent a chemical substituent of formula -RR', where R is an alkylene group of x to y carbons and R' is an aryl group as defined elsewhere herein.
  • C x-y alkheteroaryl or "C x-y alkyleneheteroaryl”
  • R is an alkylene group of x to y carbons and R" is a heteroaryl group as defined elsewhere herein.
  • Other groups preceded by the prefix “alk-” or “alkylene-” are defined in the same manner.
  • Exemplary unsubstituted alkaryl groups are of from 7 to 16 carbons.
  • alkcycloalkyl represents a cycloalkyl group attached to the parent molecular group through an alkylene group.
  • alkenyl represents monovalent straight or branched chain groups of, unless otherwise specified, from 2 to 6 carbons containing one or more carbon-carbon double bonds and is exemplified by ethenyl, 1-propenyl, 2- propenyl, 2-methyl- 1-propenyl, 1-butenyl, 2-butenyl, and the like.
  • alkheterocyclyl represents a heterocyclic group attached to the parent molecular group through an alkylene group.
  • exemplary unsubstituted alkheterocyclyl groups are of from 2 to 14 carbons.
  • alkoxy represents a chemical substituent of formula -OR, where R is an alkyl group of 1 to 6 carbons, unless otherwise specified.
  • alkoxyalkyl represents an alkyl group which is substituted with an alkoxy group.
  • exemplary unsubstituted alkoxyalkyl groups include between 2 to 12 carbons.
  • alkyl and the prefix "alk-,” as used herein, are inclusive of both straight chain and branched chain saturated groups of from 1 to 6 carbons, unless otherwise specified.
  • Alkyl groups are exemplified by methyl, ethyl, n- and iso-propyl, n-, sec-, iso- and tert-butyl, neopentyl, and the like, and may be optionally substituted with one, two, three or, in the case of alkyl groups of two carbons or more, four substituents independently selected from the group consisting of: (1) alkoxy of one to six carbon atoms; (2) alkylsulfinyl of one to six carbon atoms; (3) alkylsulfonyl of one to six carbon atoms; (4) amino; (5) aryl; (6) arylalkoxy; (7) aryloyl; (8) azido; (9) carboxaldehyde; (10) cycloalkyl
  • alkylene represents a saturated divalent hydrocarbon group derived from a straight or branched chain saturated hydrocarbon by the removal of two hydrogen atoms, and is exemplified by methylene, ethylene, isopropylene, and the like.
  • alkylsulfinyl represents an alkyl group attached to the parent molecular group through an -S(O)- group.
  • exemplary unsubstituted alkylsulfinyl groups are of from 1 to 6 carbons.
  • alkylsulfonyl represents an alkyl group attached to the parent molecular group through an -SO 2 - group.
  • exemplary unsubstituted alkylsulfonyl groups are of from 1 to 6 carbons.
  • alkylsulfinylalkyl represents an alkyl group, as defined herein, substituted by an alkylsulfinyl group.
  • exemplary unsubstituted alkylsulfinylalkyl groups are of from 2 to 12 carbons.
  • alkylsulfonylalkyl represents an alkyl group, as defined herein, substituted by an alkylsulfonyl group.
  • exemplary unsubstituted alkylsulfonylalkyl groups are of from 2 to 12 carbons.
  • alkynyl represents monovalent straight or branched chain groups of from two to six carbon atoms containing a carbon-carbon triple bond and is exemplified by ethynyl, 1-propynyl, and the like.
  • amino represents an -NH 2 group, or an -NHR N1 wherein R N1 can be a OH, NO 2 , NH 2 , NR N2 2 , SO 2 OR N2 , SO 2 R N2 , SOR N2 , and wherein R N2 can be a H, an alkyl group, or an aryl group.
  • aminoalkyl represents an alkyl group, as defined herein, substituted by an amino group.
  • aryl represents a mono- or bicyclic carbocyclic ring system having one or two aromatic rings and is exemplified by phenyl, naphthyl, 1,2- dihydronaphthyl, 1,2,3,4-tetrahydronaphthyl, fluorenyl, indanyl, indenyl, and the like, and may be optionally substituted with one, two, three, four, or five substituents independently selected from the group consisting of: (1) alkanoyl of one to six carbon atoms; (2) alkyl of one to six carbon atoms; (3) alkoxy of one to six carbon atoms; (4) alkoxyalkyl, where the alkyl and alkylene groups are independently of one to six carbon atoms; (5) alkylsulfinyl of one to six carbon atoms
  • arylalkoxy represents an alkaryl group attached to the parent molecular group through an oxygen atom.
  • exemplary unsubstituted arylalkoxy groups are of from 7 to 16 carbons.
  • aryloxy represents a chemical substituent of formula -OR', where R' is an aryl group of 6 to 18 carbons, unless otherwise specified.
  • aryloyl and “aroyl” as used interchangeably herein, represent an aryl group that is attached to the parent molecular group through a carbonyl group.
  • exemplary unsubstituted aryloyl groups are of 7 or 11 carbons.
  • azidoalkyl represents an azido group attached to the parent molecular group through an alkyl group.
  • Carboxyaldehyde represents a CHO group.
  • Carboxaldehydealkyl represents a carboxyaldehyde group attached to the parent molecular group through an alkylene group.
  • cycloalkyl represents a monovalent saturated or unsaturated non-aromatic cyclic hydrocarbon group of from three to eight carbons, unless otherwise specified, and is exemplified by cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, bicyclo[2.2.1.]heptyl and the like.
  • the cycloalkyl groups of this invention can be optionally substituted with (1) alkanoyl of one to six carbon atoms; (2) alkyl of one to six carbon atoms; (3) alkoxy of one to six carbon atoms; (4) alkoxyalkyl, where the alkyl and alkylene groups are independently of one to six carbon atoms; (5) alkylsulfinyl of one to six carbon atoms; (6) alkylsulfinylalkyl, where the alkyl and alkylene groups are independently of one to six carbon atoms; (7) alkylsulfonyl of one to six carbon atoms; (8) alkylsulfonylalkyl, where the alkyl and alkylene groups are independently of one to six carbon atoms; (9) aryl; (10) amino; (11) aminoalkyl of one to six carbon atoms; (12) heteroaryl; (13) alkaryl, where the alkylene group is of one to six carbon atoms;
  • cycloalkyloxy or "cycloalkoxy”, as used interchangeably herein, represent a cycloalkyl group, as defined herein, attached to the parent molecular group through an oxygen atom.
  • exemplary unsubstituted cycloalkyloxy groups are of from 3 to 8 carbons.
  • an "effective amount” or a "sufficient amount " of an agent, as used herein, is that amount sufficient to effect beneficial or desired results, such as clinical results, and, as such, an "effective amount” depends upon the context in which it is being applied.
  • an effective amount of an agent is, for example, an amount sufficient to achieve a reduction in NOS activity as compared to the response obtained without administration of the agent.
  • halide or "halogen” or “Hal” or “halo,” as used herein, represent bromine, chlorine, iodine, or fluorine.
  • heteroaryl represents that subset of heterocycles, as defined herein, which are aromatic: i.e., they contain 4n+2 pi electrons within the mono- or multicyclic ring system.
  • heterocycle or “heterocyclyl,” as used interchangeably herein represent a 5-, 6- or 7-membered ring, unless otherwise specified, containing one, two, three, or four heteroatoms independently selected from the group consisting of nitrogen, oxygen and sulfur.
  • the 5-membered ring has zero to two double bonds and the 6- and 7-membered rings have zero to three double bonds.
  • heterocyclyl also represents a heterocyclic compound having a bridged multicyclic structure in which one or more carbons and/or heteroatoms bridges two non- adjacent members of a monocyclic ring, e.g., a quinuclidinyl group.
  • heterocycle includes bicyclic, tricyclic and tetracyclic groups in which any of the above heterocyclic rings is fused to one, two, or three rings, e.g., an aryl ring, a cyclohexane ring, a cyclohexene ring, a cyclopentane ring, a cyclopentene ring and another monocyclic heterocyclic ring, such as indolyl, quinolyl, isoquinolyl, tetrahydroquinolyl, benzofuryl, benzothienyl and the like.
  • fused heterocycles include tropanes and 1, 2,3,5, 8,8a-hexahydroindolizine.
  • Heterocyclics include pyrrolyl, pyrrolinyl, pyrrolidinyl, pyrazolyl, pyrazolinyl, pyrazolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, pyridyl, piperidinyl, homopiperidinyl, pyrazinyl, piperazinyl, pyrimidinyl, pyridazinyl, oxazolyl, oxazolidinyl, isoxazolyl, isoxazolidiniyl, morpholinyl, thiomorpholinyl, thiazolyl, thiazolidinyl, isothiazolyl, isothiazolidinyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzothiazolyl, benzoxazolyl, furyl, thienyl, thiazolidin
  • F' is selected from the group consisting of -CH 2 -, -CH 2 O- and -O-
  • G' is selected from the group consisting of -C(O)- and -(C(R')(R")) V -
  • each of R 1 and R" is, independently, selected from the group consisting of hydrogen or alkyl of one to four carbon atoms
  • v is one to three and includes groups, such as 1,3- benzodioxolyl, 1,4-benzodioxanyl, and the like.
  • any of the heterocycle groups mentioned herein may be optionally substituted with one, two, three, four or five substituents independently selected from the group consisting of: (1) alkanoyl of one to six carbon atoms; (2) alkyl of one to six carbon atoms; (3) alkoxy of one to six carbon atoms; (4) alkoxyalkyl, where the alkyl and alkylene groups are independently of one to six carbon atoms; (5) alkylsulfinyl of one to six carbon atoms; (6) alkylsulfinylalkyl, where the alkyl and alkylene groups are independently of one to six carbon atoms; (7) alkylsulfonyl of one to six carbon atoms; (8) alkylsulfonylalkyl, where the alkyl and alkylene groups are independently of one to six carbon atoms; (9) aryl; (10) amino; (11) aminoalkyl of one to six carbon atoms; (12) heteroaryl; (13) al
  • heterocyclyloxy and “(heterocycle)oxy,” as used interchangeably herein, represent a heterocycle group, as defined herein, attached to the parent molecular group through an oxygen atom.
  • heterocyclyloyl and “(heterocycle)oyl,” as used interchangeably herein, represent a heterocycle group, as defined herein, attached to the parent molecular group through a carbonyl group.
  • hydroxy or "hydroxyl,” as used herein, represents an -OH group.
  • hydroxyalkyl represents an alkyl group, as defined herein, substituted by one to three hydroxy groups, with the proviso that no more than one hydroxy group may be attached to a single carbon atom of the alkyl group and is exemplified by hydroxymethyl, dihydroxypropyl, and the like.
  • inhibitor or “suppress” or “reduce,” as relates to a function or activity, such as NOS activity, means to reduce the function or activity when compared to otherwise same conditions except for a condition or parameter of interest, or alternatively, as compared to another condition.
  • N-protected amino refers to an amino group, as defined herein, to which is attached an N-protecting or nitrogen-protecting group, as defined herein.
  • N-protecting group and “nitrogen protecting group,” as used herein, represent those groups intended to protect an amino group against undesirable reactions during synthetic procedures. Commonly used N-protecting groups are disclosed in Greene, “Protective Groups In Organic Synthesis,” 3 rd Edition (John Wiley & Sons, New York, 1999), which is incorporated herein by reference.
  • N- protecting groups include acyl, aroyl, or carbamyl groups such as formyl, acetyl, propionyl, pivaloyl, t-butylacetyl, 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloroacetyl, phthalyl, o-nitrophenoxyacetyl, ⁇ -chlorobutyryl, benzoyl, 4- chlorobenzoyl, 4-bromobenzoyl, 4-nitrobenzoyl, and chiral auxiliaries such as protected or unprotected D, L or D, L-amino acids such as alanine, leucine, phenylalanine, and the like; sulfonyl groups such as benzenesulfonyl, p- toluenesulfonyl, and the like; carbamate forming groups such as benzyloxycarbonyl, p-
  • N-protecting groups are formyl, acetyl, benzoyl, pivaloyl, t-butylacetyl, alanyl, phenylsulfonyl, benzyl, t-butyloxycarbonyl (Boc), and benzyloxycarbonyl (Cbz).
  • nitro represents an -NO 2 group.
  • perfluoroalkyl represents an alkyl group, as defined herein, where each hydrogen radical bound to the alkyl group has been replaced by a fluoride radical.
  • Perfluoroalkyl groups are exemplified by trifluoromethyl, pentafluoroethyl, and the like.
  • perfluoroalkoxy represents an alkoxy group, as defined herein, where each hydrogen radical bound to the alkoxy group has been replaced by a fluoride radical.
  • pharmaceutically acceptable salt represents those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and animals without undue toxicity, irritation, allergic response and the like and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, S. M Berge et al. describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences 66: 1-19, 1977.
  • the salts can be prepared in situ during the final isolation and purification of the compounds of the invention or separately by reacting the free base group with a suitable organic acid.
  • Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphersulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthal
  • alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine and the like.
  • prodrugs represents those prodrugs of the compounds of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and animals with undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the invention.
  • Ph as used herein means phenyl.
  • prodrug represents compounds which are rapidly transformed in vivo to the parent compound of the above formula, for example, by hydrolysis in blood. Prodrugs of the compounds of the invention may be conventional esters.
  • esters which have been utilized as prodrugs are phenyl esters, aliphatic (C 7 -C 8 or C 8 -C 24 ) esters, cholesterol esters, acyloxymethyl esters, carbamates, and amino acid esters.
  • a compound of the invention that contains an OH group may be acylated at this position in its prodrug form.
  • nNOS selective nNOS inhibitor
  • a selective nNOS inhibitor refers to a substance that inhibits or binds the nNOS isoform more effectively than the eNOS and/or iNOS isoform as measured by an in vitro assay, such as, for example, those assays described herein.
  • Selective inhibition can be expressed in terms of an IC 50 value, a K 1 value, or the inverse of a percent inhibition value which is lower, or conversely a higher % inhibition when the substance is tested in an nNOS assay than when tested in an eNOS and/or iNOS assay.
  • the IC 50 or K value is 2 times lower. More preferably, the IC 50 or K, value is 5, 10, 50, or even more than 100 times lower.
  • solvate means a compound of the invention wherein molecules of a suitable solvent are incorporated in the crystal lattice.
  • a suitable solvent is physiologically tolerable at the dosage administered. Examples of suitable solvents are ethanol, water and the like. When water is the solvent, the molecule is referred to as a "hydrate.”
  • spirocycle represents an alkylene diradical, both ends of which are bonded to the same carbon atom of the parent group to form a spirocyclic group and also heteroalkylene diradical, both ends of which are bonded to the same atom.
  • sulfonyl represents an -S(O) 2 - group.
  • thioalkheterocyclyl represents a thioalkoxy group substituted with a heterocyclyl group.
  • thioalkoxy represents an alkyl group attached to the parent molecular group through a sulfur atom.
  • exemplary unsubstituted alkylthio groups are of from 1 to 6 carbons.
  • treatment is an approach for obtaining beneficial or desired results, such as clinical results.
  • beneficial or desired results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions; diminishment of extent of disease, disorder, or condition; stabilized (i.e. not worsening) state of disease, disorder, or condition; preventing spread of disease, disorder, or condition; delay or slowing the progress of the disease, disorder, or condition; amelioration or palliation of the disease, disorder, or condition; and remission (whether partial or total), whether detectable or undetectable.
  • Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • “Palliating" a disease, disorder, or condition means that the extent and/or undesirable clinical manifestations of the disease, disorder, or condition are lessened and/or time course of the progression is slowed or lengthened, as compared to the extent or time course in the absence of treatment.
  • the term also includes prophylactic treatment or treatment that prevents one or more symptoms or conditions of a disease, disorder, or condition.
  • Figure 1 is a schematic depiction of the Chung model of thermal hyperalgesia.
  • Figure 2 is a graph showing the dose response of the compound of Example 11 on hyperalgesia in the Chung model.
  • Figure 3 is a graph showing the effect of the compound of Example 3 on hyperalgesia in the Chung model.
  • Figure 4 is a graph showing the effect of the compound of Example 23 on hyperalgesia in the Chung model.
  • Figure 5 is a schematic depiction of the Chung model of tactile hypersensitivity.
  • Figure 6 is a graph showing the effect of the compound of Example 23 on allodynia in the Chung model.
  • Figure 7 is a schematic depiction of the Porreca model of tactile hypersensitivity.
  • Figure 8 is a graph showing the effect of the compound of Example 23 on allodynia in the Porreca model.
  • Figure 9 is a graph showing the effect of the compound of Example 37 on hyperalgesia in the Chung model.
  • Figure 10 is a graph showing the effect of the compound of Example 47 on hyperalgesia in the Chung model.
  • Figure 11 is a graph showing the effect of the compound of Example 54 on hyperalgesia in the Chung model.
  • Figure 12 is a graph showing the effect of the compound of Example 28 on hyperalgesia in the Chung model.
  • Figure 13 is a graph showing the effect of compound 28 on allodynia in the Porreca model.
  • Figure 14 is a graph showing the effect a single dose of compound 11 on allodynia in the Sciatic Nerve Cuff model.
  • Figure 15 is a graph showing the effect a multiple doses of compound 11 on allodynia in the Sciatic Nerve Cuff model.
  • Figure 16 is a graph showing the effect of compound 11 on the frequency of paw lifts in the Sciatic Nerve Cuff model.
  • Figure 17 is a graph showing the effect of compound 11 on the hind leg weight bearing in the Sciatic Nerve Cuff model.
  • Figure 18 is a schematic depiction of a model for visceral pain.
  • Figure 19 is a graph showing the effect of compound 11 in a rat model of visceral pain.
  • the invention features novel quinolones, tetrahydroquinolines, and related compounds having nitric oxide synthase (NOS) inhibitory activity, pharmaceutical and diagnostic compositions containing them, and their medical use.
  • NOS nitric oxide synthase
  • Exemplary compounds of the invention are shown in Table 2.
  • the compounds of the invention can be prepared by processes analogous to those established in the art, for example, by the reaction sequences shown in Schemes 1-8.
  • a compound of formula 3, where R 1 and Q is as defined elsewhere herein, can be prepared under standard alkylating conditions by treating a compound of formula 1 with a compound of formula 2, or a suitably protected derivative thereof, where R 1 is as defined above, with the exception that R 1 is not H, and "LG" is a leaving group, such as, for example, chloro, bromo, iodo, or sulfonate (e.g., mesylate, tosylate, or triflate).
  • Conditions to effect the alkylation of a compound of formula 1 with a compound of formula 2 may include, for example, heating a compound of formula 1 and a compound of formula 2, with or without a solvent, optionally in the presence of a suitable base (see Scheme 1).
  • Preferred conditions include, for example, heating a compound of formula 1 and 2 in the presence of a solvent, such as DMF, and potassium carbonate.
  • a compound of formula 4 can be prepared by reduction of the nitro group of a compound of formula 3 or a suitably protected derivative, under standard conditions as shown in Scheme 1.
  • standard reduction conditions include the use of Raney Nickel in a polar solvent, such as, for example, methanol or ethanol at refluxing temperatures.
  • a compound of formula 4 can be prepared by the hydrogenation of a compound of formula 3 using a suitable catalyst, such as palladium on charcoal in ethanol or another solvent or combinations of solvents.
  • a compound of formula 6 can be prepared by reacting a compound of formula 5 with a compound of formula 4 according to a previous procedure (US 200610258721 Al).
  • R 1A , R 1B , R 1C , R 1D , Y 1 is CH 2 , O, S, NR 1 , nl, pi, and ql as defined by example herein, involves the reaction of a compound of formula 7, wherein LG is a suitable leaving group, such as, for example, chloro, bromo, iodo, or sulfonate (e.g., mesylate, tosylate, or triflate), with compounds of formula 8, where X 1 is as defined above, under standard alkylation conditions as shown in Scheme 2.
  • LG is an aldehyde or ketone group
  • standard reductive amination conditions Abdel-Majid et al. J. Org. Chem.
  • a suitable reducing agent such as NaBH 4 , NaBH(OAc) 3 , NaCNBH 4 , and the like
  • an alcoholic solvent such as ethanol
  • the reductive amination may be performed in one reaction or the imine resulting from mixing a compound of formula 7 with a compound of formula 8 can be preformed in situ, followed by sequential reduction with a suitable reducing agent.
  • Compound 9 is converted to compound 11 by nitro reduction followed by amidation in a similar fashion as described in Scheme 1.
  • a suitable protecting group for example a Boc group
  • the protecting group can then be removed under standard conditions (e.g., under acidic conditions when a Boc group is used) to produce compounds of the invention.
  • R IA and/or R 1B is a methyl group
  • compounds of 9 can be treated with a demethylating agent such as a chloroformate reagent, for example, phenyl chloroformate, chloromethylchlorformate, and the like.
  • a demethylating agent such as a chloroformate reagent, for example, phenyl chloroformate, chloromethylchlorformate, and the like.
  • the protecting group can be removed by cleavage under standard conditions.
  • a preferred protecting group is the phenyl carbamate, and preferred deprotection conditions include reaction under basic conditions (e.g., sodium hydroxide) in a suitable solvent such as water in the presence of a cosolvent such as methanol or ethanol.
  • Compound of general formula 12 can be prepared from a compound of formula 4 by amide reduction with lithium aluminum hydride in aprotic solvents, such as, THF, ether, and the like.
  • a compound of formula 12 can be reduced using a suitable reducing agent, such as, BH 3 , in a suitable aprotic solvent, such as THF.
  • a suitable reducing agent such as, BH 3
  • THF a suitable aprotic solvent
  • Compounds of general formula 16 can be prepared from compound 14 and compound of general formula 15 under standard reductive amination conditions (Abdel-Majid et al. J. Org. Chem. 61:3849-3862, 1996).
  • Compounds of general formula 17 can be prepared by aromatic halogenation of compound of general formula 16 according to established procedures (de Ia Mare, "Electrophilic Halogenation," Cambridge University Press, Cambridge (1976)). The preferred condition is reacting compounds of general formula 16 with N-Bromosuccinimide under neutral conditions.
  • a compound of formula 18 can be prepared by metal catalyzed amination of a compound of formula 17 where X is chloro, bromo, or iodo (Wolfe, et al. J.
  • a suitable ammonia equivalent such as benzophenone imine, LiN(SiMe 3 ) 2 , Ph 3 SiNH 2 , NaN(SiMe 3 ) 2 , or lithium amide (Huang and Buchwald, Org. Lett. 3(21):3417-3419, 2001).
  • a preferred halogen is bromo in the presence of palladium (0) or palladium (II) catalyst.
  • suitable metal catalysts include, for example, a palladium catalyst coordinated to suitable ligands.
  • Suitable palladium catalysts include tris-dibenzylideneacetone dipalladium (Pd 2 dba 3 ) and palladium acetate (PdOAc 2 ), preferably Pd 2 dba 3 .
  • Suitable ligands for palladium can vary greatly and may include, for example, XantPhos, BINAP, DPEphos, dppf, dppb, DPPP, ( ⁇ -biphenyl)-P(t-Bu) 2 , (obi ⁇ henyl)-P(Cy) 2 , P( t -Bu)3, P(Cy) 3 , and others (Huang and Buchwald, Org. Lett. 3(21):3417-3419, 2001).
  • the ligand is P(t-Bu) 3 .
  • the Pd-catalyzed amination is performed in a suitable solvent, such as THF, dioxane, toluene, xylene, DME, and the like, at temperatures between room temperature and reflux. Conversion of compound 18 to 19 was done under conditions in Scheme 1.
  • a suitable protecting group includes a t-butoxycarbonyl (Boc) group that can be cleaved under acidic conditions, for example, aqueous HCl with an optional cosolvent.
  • R is defined herein, can be prepared by reacting compound of general formula 20 with compound of general formula 15 under standard reductive amination conditions (Abdel-Majid et al. J. Org. Chem. 61:3849-3862, 1996).
  • Compounds of general formula 23 can be prepared by cross-coupling of acrylic ester 22 with a compound of general formula 21 under Heck reaction conditions using a suitable palladium catalyst (Beletskaya and Cheprakov. Chem. Rev.100:3009-3066, 200).
  • a preferred halogen is iodo in the presence of palladium (0) or palladium (II) catalyst.
  • Suitable palladium catalysts include tris-dibenzylideneacetone dipalladium (Pd 2 dba 3 ) and palladium acetate (PdOAc 2 ), preferably PdOAc 2 .
  • Suitable ligands for palladium can vary greatly and may include, for example, (o-toZy/) 3 P XantPhos, BINAP, DPEphos, dppf, dppb, DPPP, ( ⁇ -biphenyl)-PO-Bu) 2 , (o-biphenyl)-P(Cy) 2 , P( t -Bu)3, P(Cy) 3 , and others.
  • a compound of formula 24 can be prepared by the hydrogenation of a compound of formula 23 using a suitable catalyst, such as palladium on charcoal in ethanol or another solvent or combination of solvents.
  • a suitable catalyst such as palladium on charcoal in ethanol or another solvent or combination of solvents.
  • a number of different hydrogenation conditions can also be employed in the transformation of 23 to 24 (see Rylander, "Hydrogenation Methods," Academic Press, New York (1985), Chpt 2).
  • a compound of general formula 25 can be prepared by hydrolysis of the ester 24 under standard conditions, such as, for example, aqueous sodium hydroxide in methanol. The subsequent cyclization can be performed by heating under aqueous acidic conditions (Ogawa et al J. Med.
  • a compound of formula 40 can be prepared starting from the oxindole 36 according to Scheme 8. Reacting 36 with an appropriately protected ketone of formula 15 in the presence of base, such as ammonia, pyrrolidine, and the like in a suitable solvent, such as ethanol or methanol and the like gives a compound of formula 37. Preferred conditions are ammonia in refluxing methanol.
  • a compound of formula 38 can be prepared by reduction of the double bond and nitro group under standard hydrogenation conditions, for example, Pd on carbon under an atmossphere of hydrogen or under other conditions such as transfer hydrogenation. Conversion to 39 as described previously herein and deprotection under standard conditions provide compounds of formula 40.
  • a preferred protecting group is the Boc protecting group, which can be cleaved under acidic conditions, such as refluxing in methanolic HCl solution.
  • the chemistries outlined above may have to be modified, for instance, by the use of protective groups to prevent side reactions due to reactive groups, such as reactive groups attached as substituents. This may be achieved by means of conventional protecting groups as described in "Protective Groups in Organic Chemistry,” McOmie, Ed., Plenum Press, 1973 and in Greene and Wuts, "Protective Groups in Organic Synthesis,” John Wiley & Sons, 3 rd Edition, 1999.
  • the compounds of the invention, and intermediates in the preparation of the compounds of the invention may be isolated from their reaction mixtures and purified (if necessary) using conventional techniques, including extraction, chromatography, distillation, and recrystallization.
  • a desired compound salt is achieved using standard techniques.
  • the neutral compound is treated with an acid in a suitable solvent and the formed salt is isolated by filtration, extraction, or any other suitable method.
  • solvates of the compounds of the invention will vary depending on the compound and the solvate.
  • solvates are formed by dissolving the compound in the appropriate solvent and isolating the solvate by cooling or adding an antisolvent.
  • the solvate is typically dried or azeotroped under ambient conditions.
  • Preparation of an optical isomer of a compound of the invention may be performed by reaction of the appropriate optically active starting materials under reaction conditions which will not cause racemization.
  • the individual enantiomers may be isolated by separation of a racemic mixture using standard techniques, such as, for example, fractional crystallization or chiral HPLC.
  • a radiolabeled compound of the invention may be prepared using standard methods known in the art.
  • tritium may be incorporated into a compound of the invention using standard techniques, such as, for example, by hydrogenation of a suitable precursor to a compound of the invention using tritium gas and a catalyst.
  • a compound of the invention containing radioactive iodine may be prepared from the corresponding trialkyltin (suitably trimethyltin) derivative using standard iodination conditions, such as [ 125 I] sodium iodide in the presence of chloramine-T in a suitable solvent, such as dimethylformamide.
  • the trialkyltin compound may be prepared from the corresponding non-radioactive halo, suitably iodo, compound using standard palladium-catalyzed stannylation conditions, such as, for example, hexamethylditin in the presence of tetrakis(triphenylphosphine) palladium (0) in an inert solvent, such as dioxane, and at elevated temperatures, suitably 50-100 0 C.
  • standard palladium-catalyzed stannylation conditions such as, for example, hexamethylditin in the presence of tetrakis(triphenylphosphine) palladium (0) in an inert solvent, such as dioxane, and at elevated temperatures, suitably 50-100 0 C.
  • the present invention features all uses for compounds of the invention, including use in therapeutic methods, whether alone or in combination with another therapeutic substance, their use in compositions for inhibiting NOS activity, e.g., nNOS, their use in diagnostic assays, and their use as research tools.
  • NOS activity e.g., nNOS
  • the compounds of the invention have useful NOS inhibiting activity, and therefore are useful for treating, or reducing the risk of, diseases or conditions that are ameliorated by a reduction in NOS activity.
  • diseases or conditions include those in which the synthesis or oversynthesis of nitric oxide plays a contributory part.
  • the present invention features a method of treating, or reducing the risk of, a disease or condition caused by NOS activity that includes administering an effective amount of a compound of the invention to a cell or animal in need thereof.
  • Such diseases or conditions include, for example, migraine headache (with or without aura), chronic tension type headache (CTTH), migraine with allodynia, medication overuse headache, neuropathic pain, AIDS associated painful neuropathy, chronic headache, central post-stroke pain (CPSP), medication-induced hyperalgesia and/or allodynia, e.g., opioid-induced hyperalgesia or triptan (5-HT1D/1B agonists)- induced hyperalgesia/allodynia, acute pain, chronic pain, diabetic neuropathy, trigeminal neuralgia, chemotherapy induced neuropathic pain (e.g., Taxol, cis-Platin, Doxorubicin etc.), bone cancer pain, chemical dependencies or addictions, e.g., drug addiction, cocaine addition, nicotine addition, metamphetamine-induced neurotoxicity, ethanol tolerance, dependence, or withdrawal, or morphine/opioid induced tolerance, dependence, hyperalgesia, or withdrawal, CNS disorders including but not limited
  • late sumatriptan intervention in rats did not prevent I.S.- induced neuronal sensitivity to mechanical stimulation at the periorbital skin, nor decreased the threshold to heat (a clinical correlate of patients with mechanical and thermal allodynia in the periorbital area).
  • early sumatriptan prevented LS. from inducing both thermal and mechanical hypersensitivity.
  • late sumatriptan intervention reverses the enlargement of dural receptive fields and increases in sensitivity to dural indentation (a clinical correlate of pain throbbing exacerbated by bending over) while early intervention prevents its development.
  • triptans were effective in terminating the throbbing of migraine whether administered early or late, the peripheral action of sumatriptan is unable to terminate migraine pain with allodynia following late intervention via the effects of central sensitization of trigeminovascular neurons.
  • the limitations of triptans suggest that improvement in the treatment of migraine pain can be achieved by utilizing drugs that can abort ongoing central sensitization, such as the compounds of the present invention.
  • CTH Chronic Headache
  • NMMA an NOS inhibitor
  • the NOS inhibitors of the present invention may be useful for the treatment of chronic tension-type headache.
  • NO evokes pain on intracutaneous injection (Holthusen and Arndt, Neurosci. Lett. 165:71-74, 1994), thus showing a direct involvement of NO in pain. Furthermore, NOS inhibitors have little or no effect on nociceptive transmission under normal conditions (Meller and Gebhart, Pain 52:127-136, 1993). NO is involved in the transmission and modulation of nociceptive information at the periphery, spinal cord and supraspinal level (Duarte et al., Eur. J. Pharmacol. 217:225-227, 1992; Haley et al., Neuroscience 31:251-258, 1992).
  • NOS inhibitors 7-NI and L-NAME relieve chronic allodynia-like symptoms in rats with spinal cord injury (Hao and Xu, Pain 66:313-319, 1996).
  • the effects of 7-NI were not associated with a significant sedative effect and were reversed by L-arginine (NO precursor).
  • the maintenance of thermal hyperalgesia is believed to be mediated by nitric oxide in the lumbar spinal cord and can be blocked by intrathecal administration of a nitric oxide synthase inhibitor like L-NAME or soluble guanylate cyclase inhibitor methylene blue ⁇ Neuroscience 50(l):7-10, 1992).
  • the NOS inhibitors of the present invention may be useful for the treatment of chronic or neuropathic pain.
  • the endogenous polyamine metabolite agmatine is a metabolite of arginine that is both an NOS inhibitor and N-methyl-D-aspartate (NMDA) channel antagonist.
  • Agmatine is effective in both the spinal nerve ligation (SNL) model of neuropathic pain as well as the streptozotocin model of diabetic neuropathy (Karadag et al., Neurosci. Lett. 339(l):88-90, 2003).
  • SNL spinal nerve ligation
  • streptozotocin model of diabetic neuropathy Karadag et al., Neurosci. Lett. 339(l):88-90, 2003.
  • compounds possessing NOS inhibitory activity such as, for example, a compound of formula I, a combination of an NOS inhibitor and an NMDA antagonist should be effective in treating diabetic neuropathy and other neuropathic pain conditions.
  • LPS a well-known pharmacological tool, induces inflammation in many tissues and activates NFKB in all brain regions when administered intravenously. It also activates pro-inflammatory genes when injected locally into the striatum (Stern et al., 7. Neuroimmunology, 109:245-260, 2000). Recently it has been shown that both the NMDA receptor antagonist MK801 and the brain selective nNOS inhibitor 7-NI both reduce NFKB activation in the brain and thus reveal a clear role for glutamate and NO pathway in neuroinflammation (Glezer et al., Neuropharmacology
  • NO in cerebral ischemia can be protective or destructive depending on the stage of evolution of the ischemic process and on the cellular compartment producing NO (Dalkara et al., Brain Pathology 4:49, 1994). While the NO produced by eNOS is likely beneficial by acting as a vasodilator to improve blood flow to the affected area (Huang et al., J. Cereb. Blood Flow Metab. 16:981, 1996), NO produced by nNOS contributes to the initial metabolic deterioration of the ischemic penumbra, resulting in larger infarcts (Hara et al., J. Cereb. Blood Flow Metab. 16:605, 1996).
  • the metabolic derangement that occurs during ischemia and subsequent reperfusion results in the expression and release of several cytokines that activate iNOS in several cell types including some of the central nervous system.
  • NO can be produced at cytotoxic levels by iNOS, and increased levels of iNOS contribute to progressive tissue damage in the penumbra, leading to larger infarcts (Parmentier et al., Br. J. Pharmacol. 127:546, 1999). Inhibition of i-NOS has been shown to ameliorate cerebral ischemic damage in rats (Am. J. Physiol. 268:R286, 1995).
  • NMDA antagonist e.g., MK-801 or LY293558
  • nNOS selective inhibitors 7-NI or ARLl 7477
  • the compounds of the invention administered either alone or in combination with NMDA antagonists, or compounds possessing mixed nNOS/NMDA activity, may be effective in treating conditions of stroke and other neurodegenerative disorders.
  • Cerebral damage and cognitive dysfunction still remains as a major complication of patients undergoing coronary artery bypass surgery (CABG) (Roch et al., N. Eng. J. Med. 335:1857-1864, 1996; Shaw et al., Q. J. Med. 58:59-68, 1986).
  • CABG coronary artery bypass surgery
  • This cerebral impairment following surgery is a result of ischemia from preoperative cerebral microembolism.
  • ⁇ MDA antagonist remacemide patients showed a significant overall postoperative improvement in learning ability in addition to reduced deficits (Arrowsmith et al., Stroke 29:2357-2362, 1998).
  • a neuroprotective agent such as a compound of the invention or an ⁇ MDA antagonist, either alone or in combination (as discussed above), may have a beneficial effect improving neurological outcomes after CABG.
  • HIV-I infection can give rise to dementia.
  • the HIV-I coat protein gp-120 kills neurons in primary cortical cultures at low picomolar levels and requires external glutamate and calcium (Dawson et al., 90(8):3256-3259, 1993).
  • This toxicity can be attenuated by administration of a neuroprotective agent, e.g., a compound of the invention, either alone or in combination with another therapeutic agent, such as, for example, an ⁇ MDA antagonist (as discussed above).
  • Cardiogenic Shock Cardiogenic shock is the leading cause of death for patients with acute myocardial infarction that is consistent with increased levels of NO and inflammatory cytokines.
  • High levels of NO and peroxynitrite have many effects, including a direct inhibition on myocardial contractability, suppression of mitochondrial respiration in myocardium, alteration in glucose metabolism, reduced catacholamine responsivity, and induction of systemic vasodilation (Hochman, Circulation 107:2998, 2003).
  • administration of the NOS inhibitor L-NMMA resulted in increases in urine output and blood pressure and survival rate of 72% up to 30 days (Cotter et al., Circulation 101:1258-1361, 2000).
  • NOS inhibitors have antidepressant activity in mice (Harkin et al. Eur. J. Pharm. 372:207-213, 1999) and that their effect is mediated by a serotonin dependent mechanism (Harkin et ⁇ ., Neuropharmacology 44(5):6l ⁇ -623, 1993). 7-NI demonstrates anxiolytic activity in the rat plus-maze test (Yildiz et al., Pharmacology, Biochemistry and Behavior 65: 199-202, 2000), whereas the selective nNOS inhibitor TRIM is effective in both the FST model of depression and anxiety in the light-dark compartment test (Volke et al., Behavioral Brain Research 140(1-2): 141-7, 2003).
  • Administration of a compound of the invention to an afflicted individual, either alone or in combination with another therapeutic agent, such as, for example, an antidepressant, may be useful for the treatment of anxiety or depression. Attention Deficit Hyperactivity Disorder
  • Non-selective attention (NSA) to environmental stimuli in Spontaneously Hypertensive (SHR) and Naples Low-Excitability (NHE) rats has been used as an animal model of Attention-Deficit Hyperactivity Disorder (ADHD) (Aspide et al., Behav. Brain Res. 95(l):23-33, 1998). These genetically altered animals show increased episodes of rearing that have a shorter duration than observed in normal animals. A single injection of L-NAME at 10 mg/kg produced an increase in rearing duration. Similarly, using the more neuronally selective 7-NINA, an increase in the rearing duration was observed after rapid administration (i.p.), while a slow release single release dose or a slow multiple release dose (s.c. in DMSO) resulted in the opposite effect. Thus, administration of a compound of the invention may be useful for the treatment of ADHD.
  • ADHD Attention-Deficit Hyperactivity Disorder
  • PCP Psychiasis Phencyclidine
  • Hypothermic cardiac arrest is a technique used to protect from ischemic damage during cardiac surgery when the brain is sensitive to damage during the period of blood flow interruption.
  • Various neuroprotective agents have been used as adjunct agents during HCA and reducing nitric oxide production during HCA is predicted to result in improvements in neurological function. This is based on previous studies that showed glutamate excitotoxicity plays a role in HCA-induced neurologic damage (Redmond et al., J. Thorac. Cardiovasc. Surg. 107:776-87, 1994; Redmond et al., Ann. Thorac. Surg. 59:579-84, 1995) and that NO mediates glutamate excitotoxicity (Dawson and Snyder, J. Neurosci.
  • a neuronal NOS inhibitor was shown to reduce cerebral NO production, significantly reduce neuronal necrosis, and resulted in superior neurologic function relative to controls (Tseng et al., Ann. Thorac. Surg. 67:65-71, 1999).
  • Administration of a compound of the invention may also be useful for protecting patients from ischemic damage during cardiac surgery.
  • Mitochondrial dysfunction, glutamate excitotoxicity, and free radical induced oxidative damage appear to be the underlying pathogenesis of many neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD), Alzheimer's disease (AD), and Huntington's disease (HD) (Schulz et al., MoI Cell. Biochem. 174(1-2): 193- 197, 1997; Beal, Ann. Neurol. 38:357-366, 1995), and NO is a primary mediator in these mechanisms.
  • ALS amyotrophic lateral sclerosis
  • PD Parkinson's disease
  • AD Alzheimer's disease
  • HD Huntington's disease
  • AD The pathology of AD is associated with ⁇ -amyloid plaques infiltrated with activated microglia and astrocytes.
  • beta- amyloid When cultured rat microglia are exposed to beta- amyloid, there is a prominent microglial release of nitric oxide, especially in the presence of gamma-interferon (Goodwin et al., Brain Research 692(l-2):207-14, 1995).
  • treatment with nitric oxide synthase inhibitors provides neuroprotection against toxicity elicited by human beta-amyloid (Resink et al., Neurosci. Abstr. 21 :1010, 1995).
  • Alzheimer' s-type dementia the NMDA antagonist memantine was associated with improved Clinical Global Impression of Change and Behavioral Rating Scale for Geriatric Patients scores (Winblad and Poritis, Int. J. Geriatr. Psychiatry 14: 135-46, 1999). These results suggest that administration of an appropriate dose of an NOS inhibitor, such as, for example, a compound of the invention, can be beneficial in the treatment of AD.
  • an NOS inhibitor such as, for example, a compound of the invention
  • ALS Amyotrophic lateral sclerosis
  • HD Huntington's disease
  • Oxidative damage is one of the major consequences of defects in energy metabolism and is present in HD models after injection of excitotoxins and mitochondrial inhibitors (A. Petersen et. al., Exp. Neurol. 157:1-18, 1999). This mitochondrial dysfunction is associated with the selective and progressive neuronal loss in HD (Brown et al., Ann. Neurol. 41:646-653, 1997).
  • NO can directly impair the mitochondrial respiratory chain complex IV (Calabrese et al., Neurochem. Res. 25:1215-41, 2000). Striatal medium spiny neurons appear to be the primary target for the generation of motor dysfunction in HD. Hyperphosphorylation and activation of NMDA receptors on these neurons likely participates in the generation of motor dysfunction. It has been shown clinically that the NMDA antagonist amantadine improves choreiform dyskinesias in HD (Verhagen Metman et al., Neurology 59:694- 699, 2002).
  • nNOS inhibitors especially those with mixed nNOS/NMDA, or combinations of drugs with nNOS and NMDA activity will also be useful in ameliorating the effects and or progression of HD.
  • pretreatment of rats with 7-nitroindazole attenuates the striatal lesions elicited by stereotaxic injections of malonate, an injury that leads to a condition resembling Huntington's disease (Hobbs et. al., Ann. Rev. Pharm. Tox. 39:191-220, 1999).
  • mice at 11, 19 and 35 weeks show a progressive increase in lipid peroxidation with normal levels of superoxide dismutase (SOD) at 11 weeks similar to wild type (WT) mice; a maximum level at 19 weeks, above that observed in WT mice and corresponding to the early phase of disease progression; and finally, decreasing levels at 35 weeks below that observed in WT mice (Perez- Sevriano et al., Brain Res. 951:36-42, 2002).
  • SOD superoxide dismutase
  • levels of calcium dependent NOS was the same for 11 week mice in both WT and R6/1 mice, but increased significantly at 19 weeks and decreased at 35 weeks relative to WT control mice.
  • Levels of nNOS expression also increased dramatically relative to controls at 19 weeks but were decreased significantly relative to controls at 35 weeks. No significant differences were observed in levels of eNOS expression, nor could iNOS protein be detected during progression of the disease.
  • the progressive phenotypic expression of the disease as measured by increased weight loss, feet clasping behavior, and horizontal and vertical movements, are consistent with changes in NOS activity and nNOS expression.
  • Methamphetamine-Induced Neurotoxicity Methamphetamine is neurotoxic by destroying dopamine nerve terminals in vivo. It has been shown that methamphetamine-induced neurotoxicity can be attenuated by treatment with NOS inhibitors in vitro (Sheng et al., Ann. NY. Acad. Sci. 801: 174-186, 1996) and in in vivo animal models (Itzhak et al., Neuroreport 11(13):2943-6, 2000).
  • the nNOS selective inhibitor AR-17477AR at 5 mg/kg s.c in mice, was able to prevent the methamphetamine-induced loss of the neurofilament protein NF68 in mouse brain and prevent the loss of striatal dopamine and homovanillic acid (HVA) (Sanchez et al., J. Neurochem. 85(2):515-524, 2003).
  • HVA homovanillic acid
  • a compound of the invention may be useful for the protection or treatment of any of the neurodegenerative diseases described herein. Further, the compounds of the invention may be tested in standard assays used to assess neuroprotection (see for example, Am. J. Physiol. 268:R286, 1995).
  • L-NAME has been shown to significantly alter cocaine reinforcement by decreasing the amount of self- administration and by increasing the inter-response time between successive cocaine injections (Pudiak and Bozarth, Soc. Neurosci. Abs. 22:703, 1996). This indicates that NOS inhibition by compounds of the invention may be useful in the treatment of cocaine addiction.
  • nNOS antisense Naassila et. al., Pharmacol. Biochem. Behav. 67:629-36, 2000
  • NOS inhibition by compounds of the invention may be useful in the treatment of ethanol tolerance and dependence.
  • Administration of a compound of the invention may be useful for the treatment of chemical dependencies and drug addictions.
  • an NOS inhibitor such as, for example, a compound of the invention, either alone or in combination with another therapeutic agent, such as, for example, an antiepileptic agent, may be useful for the treatment of epilepsy or a similar disorder.
  • antiepileptic agents useful in a combination of the invention include carbamazepine, gabapentin, lamotrigine, oxcarbazepine, phenyloin, topiramate, and valproate.
  • Diabetic Nephropathy Urinary excretion of NO byproducts is increased in diabetic rats after streptozotocin treatment and increased NO synthesis has been suggested to be involved in diabetic glomerular hyperfiltration.
  • the neuronal isoform nNOS is expressed in the loop of Henle and mucula densa of the kidney and inhibition of this isoform using 7-NI reduces glomerular filtration without affecting renal arteriole pressure or renal blood flow (Sigmon et al., Gen. Pharmacol. 34(2):95-100, 2000). Both the non-selective NOS inhibitor L-NAME and the nNOS selective 7-NI normalize renal hyperfiltration in diabetic animals (Ito et al., 7. Lab Clin. Med. 138(3):177-185, 2001). Therefore, administration of a compound of the invention may be useful for the treatment of diabetic nephropathy.
  • Medication overuse headache is associated with excessive use of combination analgesics, opioids, barbiturates, aspirin, NSAIDS, caffeine and triptans and is a common problem that limits the usefulness of these types of medications (Diener and Limmroth. Medication-overuse headache: a worldwide problem. Lancet Neurol. 2004: 3, 475-483). It is generally defined as headaches that present >15 days per month (Headache Classification Committee. The International Classification of Headache Disorders (2 nd Ed). Cephalalgia 2004: 24 (Supple.1); 9-160).
  • Changes in synaptic plasticity involve changes in intracellular calcium and nitric oxide levels. Patients suffering from chronic headache, migraine and MOH patients show increased levels of platelet nitrate levels. Thus the development of the sensitization in MOH is likely mediated by the changes in NO and calcium levels in the CNS (Sarchielli et. al. Nitric oxide pathway, Ca2+, and serotonin content in platelets from patients suffering from chronic daily headache. Cephalalgia 1999: 19; 810-816). Given that the development of central sensitization is mediated by nNOS (Cizkova et. al. Brain. Res. Bull. 2002; 58(2): 161-171, Choi et. al. J. Neurol.
  • neuronal nitric oxide synthase inhibitors such as the compounds of the invention, will be useful in the prevention and treatment of MOH if used in concomitantly with other headache medications. It is also expected that both CTTH and migraine treatment with nNOS inhibitors will not result in the development of MOH.
  • nNOS constitutes more than 90% of the total NOS in the small intestine. Although iNOS is constitutively present, it accounts for less than 10% of the total NOS activity, and eNOS is essentially undetectable in the intestine (Qu XW et. al. Type I nitric oxide synthase (NOS) is the predominant NOS in rat small intestine.
  • nNOS nerve growth factor
  • the main function of nNOS in the intestine is believed to be regulation of gut motility via neuronal signal transmission in the NANC components of the nervous system. NO regulates the muscle tone of the sphincter in the lower esophagus, pylorus, sphincter of Oddi, and anus. NO also regulates the accommodation reflex of the fundus and the peristaltic reflex of the intestine. NOS inhibitors are known to delay gastric emptying and colonic transit (T. Takahashi J. Gastroenterol. 2003;38(5):421-30).
  • nNOS inhibitors can be therapeutic in GI disorders that would benefit from the delay of gastric emptying or slowing of colonic transit.
  • Dumping syndrome is a disorder that in which food is emptied too quickly from the stomach, filling the small intestine with undigested food that is not adequately prepared to permit efficient absorption of nutrients in the small intestine and is often observed after gastrectomy. Therefore, administration of a compound of the invention may be useful for the treatment of gastrointestinal disorders such as dumping syndrome.
  • one or more compounds of the invention can be used in combination with other therapeutic agents.
  • one or more compounds of the invention can be combined with another NOS inhibitor.
  • Exemplary inhibitors useful for this purpose include, without limitation, those described in U.S. Patent No. 6,235,747; U.S. Patent Applications Serial Nos. 09/127,158, 09/325,480, 09/403,177, 09/802,086, 09/826,132, 09/740,385, 09/381,887, 10/476,958, 10/483,140, 10/484,960, 10/678,369, 10/819,853, 10/938,891; International Publication Nos.
  • one or more compounds of the invention can be combined with an antiarrhythmic agent.
  • antiarrhythmic agents include, without limitation, lidocaine and mixiletine.
  • GABA-B agonists, alpha-2-adrenergic receptor agonists, cholecystokinin antagonists, 5HT 1B/1D agonists, or CGRP antagonists can also be used in combination with one or more compounds of the invention.
  • alpha-2- adrenergic receptor agonists include clonidine, lofexidine, and propanolol.
  • Non- limiting examples of cholecystokinin antagonists include L-365, 260; CI-988; LY262691; S0509, or those described in U.S. Patent No. 5,618,811.
  • Non-limiting examples of 5HT 1B/ID agonists that may be used in combination with a compound of the invention include dihydroegotamine, eletriptan, frovatriptan, naratriptan, rizatriptan, sumatriptan, donitriptan, or zolmitriptan.
  • Non-limiting examples of CGRP antagonists that may be used in combination with a compound of the invention include quinine analogues as described in International Publication No. WO9709046, non-peptide antagonists as described in International Publication Nos. WOOl 32648, WO0132649, WO9811128, WO9809630, WO9856779, WO0018764, or other antagonists such as SB-(+)-273779 or BIBN-4096BS.
  • Substance P antagonists also known as NKi receptor antagonists
  • NKi receptor antagonists are also useful in combination with one or more compounds of the invention.
  • Exemplary inhibitors useful for this purpose include, without limitation, those compounds disclosed in U.S. Patent Nos. 3,862,114, 3,912,711, 4,472,305, 4,481,139, 4,680,283, 4,839,465, 5,102,667, 5,162,339, 5,164,372, 5,166,136, 5,232,929, 5,242,944, 5,300,648, 5,310,743, 5,338,845, 5,340,822, 5,378,803, 5,410,019, 5,411,971, 5,420,297, 5,422,354, 5,446,052, 5,451,586, 5,525,712, 5,527,811, 5,536,737, 5,541,195, 5,594,022, 5,561,113, 5,576,317, 5,604,247, 5,624,950, and 5,635,510; International Publication Nos.
  • Suitable classes of antidepressant agents that may be used in combination with a compound of the invention include, without limitation, norepinephrine re-uptake inhibitors, selective serotonin re-uptake inhibitors (SSRIs), selective noradrenaline/norepinephrine reuptake inhibitors (NARIs), monoamine oxidase inhibitors (MAOs), reversible inhibitors of monoamine oxidase (RIMAs), dual serotonin/noradrenaline re-uptake inhibitors (SNRIs), ⁇ -adrenoreceptor antagonists, noradrenergic and specific serotonergic antidepressants (NaSSAs), and atypical antidepressants.
  • norepinephrine re-uptake inhibitors include selective serotonin re-uptake inhibitors (SSRIs), selective noradrenaline/norepinephrine reuptake inhibitors (NARIs), monoamine oxidase inhibitors
  • Non-limiting examples of norepinephrine re-uptake inhibitors include tertiary amine tricyclics and secondary amine tricyclics, such as, for example, adinazolam, amineptine, amoxapine, butriptyline, demexiptiline, desmethylamitriptyline, desmethylclomipramine, demexiptiline, desipramine, doxepin, dothiepin, fluacizine, imipramine, imipramine oxide, iprindole, lofepramine, maprotiline, melitracen, metapramine, norclolipramine, nortriptyline, noxiptilin, opipramol, perlapine, pizotifen, pizotyline, propizepine, protriptyline, quinupramine, tianeptine, trimipramine, trimipramineamiltriptylinoxide, and pharmaceutically acceptable salts thereof.
  • Non-limiting examples of selective serotonin re-uptake inhibitors include, for example, clomipramine, femoxetine, fluoxetine, fluvoxamine, paroxetine, and sertraline, and pharmaceutically acceptable salts thereof.
  • Non-limiting examples of selective noradrenaline/norepinephrine reuptake inhibitors include, for example, atomoxetine, bupropion; reboxetine, tomoxetine, and viloxazine and pharmaceutically acceptable salts thereof.
  • Non-limiting examples of selective monoamine oxidase inhibitors include, for example, isocarboxazid, phenezine, tranylcypromine and selegiline, and pharmaceutically acceptable salts thereof.
  • Other monoamine oxidase inhibitors useful in a combination of the invention include clorgyline, cimoxatone, befloxatone, brofaromine, apelinaprine, B W-616U (Burroughs Wellcome), BW- 1370U87 (Burroughs Wellcome), CS-722 (RS-722) (Sankyo), E-2011 (Eisai), harmine, harmaline, moclobemide, PharmaProjects 3975 (Hoechst), RO 41-1049 (Roche), RS- 8359 (Sankyo), T-794 (Tanabe Seiyaku), toloxatone, K-Y 1349 (Kalir and Youdim), LY-51641 (Lilly), LY-121768 (
  • Non-limiting examples of dual serotonin/norepinephrine reuptake blockers include, for example, duloxetine, milnacipran, mirtazapine, nefazodone, and venlafaxine.
  • Non-limiting examples of other antidepressants that may be used in a method of the present invention include adinazolam, alaproclate, amineptine, amitriptyline amitriptyline/chlordiazepoxide combination, atipamezole, azamianserin, apelinaprine, befuraline, bifemelane, binodaline, bipenamol, brofaromine, caroxazone, cericlamine, cianopramine, cimoxatone, citalopram, clemeprol, clovoxamine, dazepinil, deanol, demexiptiline, dibenzepin, dimetacrine, dothiepin, d
  • opioids can be used in combination with one or more compounds of the invention.
  • opioids useful for this purpose include, without limitation, alfentanil, butorphanol, buprenorphine, dextromoramide, dezocine, dextropropoxyphene, codeine, dihydrocodeine, diphenoxylate, etorphine, fentanyl, hydrocodone, hydromorphone, ketobemidone, loperamide, levorphanol, levomethadone, meperidine, meptazinol, methadone, morphine, morphine-6- glucuronide, nalbuphine, naloxone, oxycodone, oxymorphone, pentazocine, pethidine, piritramide, propoxylphene, remifentanil, sulfentanyl, tilidine, and tramadol
  • anti-inflammatory compounds such as steroidal agents or non-steroidal anti-inflammatory drugs (NSAIDs)
  • NSAIDs non-steroidal anti-inflammatory drugs
  • steroidal agents include prednisolone and cortisone.
  • Non-limiting examples of NSAIDs include acemetacin, aspirin, celecoxib, deracoxib, diclofenac, diflunisal, ethenzamide, etofenamate, etoricoxib, fenoprofen, flufenamic acid, flurbiprofen, lonazolac, lornoxicam, ibuprofen, indomethacin, isoxicam, kebuzone, ketoprofen, ketorolac, naproxen, nabumetone, niflumic acid, sulindac, tolmetin, piroxicam, meclofenamic acid, mefenamic acid, meloxicam, metamizol, mofebutazone, oxyphenbutazone, parecoxib, phenidine, phenylbutazone, piroxicam, propacetamol, propyphenazone, rofecoxib, salicylamide, suprofen
  • any of the above combinations can be used to treat any appropriate disease, disorder, or condition.
  • Exemplary uses for combinations of a compound of the invention and another therapeutic agent are described below.
  • Nerve injury can lead to abnormal pain states known as neuropathic pain.
  • Some of the clinical symptoms include tactile allodynia (nociceptive responses to normally innocuous mechanical stimuli), hyperalgesia (augmented pain intensity in response to normally painful stimuli), and spontaneous pain.
  • Spinal nerve ligation (SNL) in rats is an animal model of neuropathic pain that produces spontaneous pain, allodynia, and hyperalgesia, analogous to the clinical symptoms observed in human patients (Kim and Chung, Pain 50:355-363, 1992; Seltzer, Neurosciences 7:211-219, 1995).
  • Neuropathic pain can be particularly insensitive to opioid treatment (Benedetti et al., Pain 74:205-211, 1998) and is still considered to be relatively refractory to opioid analgesics (MacFarlane et al., Pharmacol. Ther. 75:1-19, 1997; Watson, Clin. J. Pain 16:S49-S55, 2000). While dose escalation can overcome reduced opioid effectiveness, it is limited by increased side effects and tolerance. Morphine administration is known to activate the NOS system, which limits the analgesic action of this drug (Machelska et al., NeuroReport 8:2743-2747, 1997; Wong et al., Br. J. Anaesth.
  • opioids are an important therapy for the treatment of moderate to severe pain, in addition to the usual side effects that limit their utility, the somewhat paradoxical appearance of opioid-induced hyperalgesia may actually render patients more sensitive to pain and potentially aggravate their pain (Angst and Clark, Anesthesiology, 2006, 104(3), 570-587; Chu et. al. J. Pain 2006, 7(1) 43-48).
  • the development of tolerance and opioid induced hyperalgesia is consistent with increased levels of NO production in the brain.
  • the reduced analgesic response to opioids is due to an NO-induced upregulated hyperalgesic response (Heinzen and Pollack, Brain Res. 2004, 1023, 175-184).
  • an nNOS inhibitor with an opioid can enhance opioid analgesia in neuropathic pain and prevent the development of opioid tolerance and opioid-induced hyperalgesia.
  • neuropathic pain Pain 68:217-227, 1996) and migraine (Tomkins et al., Am. J. Med. 111:54-63, 2001), and act via the serotonergic or noradrenergic system. NO serves as a neuromodulator of these systems (Garthwaite and Boulton, Annu. Rev. Physiol. 57:683, 1995). 7-NI has been shown to potentiate the release of noradrenaline (NA) by the nicotinic acetylcholine receptor agonist DMPP via the NA transporter (Kiss et al., Neuroscience Lett. 215: 115-118, 1996).
  • NA noradrenaline
  • antidepressants such as paroxetine, tianeptine, and imipramine decrease levels of hippocampal NO (Wegener et al., Brain Res. 959:128-134, 2003). It is likely that NO is important in the mechanism by which antidepressants are effective for treating pain and depression, and that a combination of an nNOS inhibitor with an antidepressant, such as, for example, those combinations described above, will produce better treatments.
  • GTN Glyceryl trinitrate
  • CGRP Calcitonin Gene Related Peptide
  • Sumatriptan an antimigraine drug having affinity at 5HT 1B , 5HT 1D , and 5HT 1 preceptors, reduces GTN-induced immediate headache and in parallel contracts cerebral and extracerebral arteries (Iversen and Olesen, Cephalagia 13(Suppl 13): 186, 1993).
  • the antimigraine drug rizatriptan also reduces plasma levels of CGRP following migraine pain reduction (Stepien et al., Neurol. Neurochir. Pol. 37(5): 1013-23, 2003). Both NO and CGRP have therefore been implicated as a cause for migraine.
  • Serotonin 5HT 1B/1D agonists have been shown to block NMDA receptor-evoked NO signaling in brain cortex slices (Strosznajder et al., Cephalalgia 19(10):859, 1999). These results suggest that a combination of a compound of the invention and a selective or nonselective 5HT 1B/1D/1F agonist or a CGRP antagonist, such as those combinations described above, would be useful for the treatment of migraine.
  • the compounds of the invention are preferably formulated into pharmaceutical compositions for administration to human subjects in a biologically compatible form suitable for administration in vivo. Accordingly, in another aspect, the present invention provides a pharmaceutical composition comprising a compound of the invention in admixture with a suitable diluent, carrier, or excipient.
  • the compounds of the invention may be used in the form of the free base, in the form of salts, solvates, and as prodrugs. All forms are within the scope of the invention.
  • the described compounds or salts, solvates, or prodrugs thereof may be administered to a patient in a variety of forms depending on the selected route of administration, as will be understood by those skilled in the art.
  • the compounds of the invention may be administered, for example, by oral, parenteral, buccal, sublingual, nasal, rectal, patch, pump, or transdermal administration and the pharmaceutical compositions formulated accordingly.
  • Parenteral administration includes intravenous, intraperitoneal, subcutaneous, intramuscular, transepithelial, nasal, intrapulmonary, intrathecal, rectal, and topical modes of administration. Parenteral administration may be by continuous infusion over a selected period of time.
  • a compound of the invention may be orally administered, for example, with an inert diluent or with an assimilable edible carrier, or it may be enclosed in hard or soft shell gelatin capsules, or it may be compressed into tablets, or it may be incorporated directly with the food of the diet.
  • a compound of the invention may be incorporated with an excipient and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • a compound of the invention may also be administered parenterally. Solutions of a compound of the invention can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, DMSO and mixtures thereof with or without alcohol, and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms. Conventional procedures and ingredients for the selection and preparation of suitable formulations are described, for example, in Remington's Pharmaceutical Sciences (2003 - 20th edition) and in The United States Pharmacopeia: The National Formulary (USP 24 NF19), published in 1999.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases the form must be sterile and must be fluid to the extent that may be easily administered via syringe.
  • compositions for nasal administration may conveniently be formulated as aerosols, drops, gels, and powders.
  • Aerosol formulations typically include a solution or fine suspension of the active substance in a physiologically acceptable aqueous or non-aqueous solvent and are usually presented in single or multidose quantities in sterile form in a sealed container, which can take the form of a cartridge or refill for use with an atomizing device.
  • the sealed container may be a unitary dispensing device, such as a single dose nasal inhaler or an aerosol dispenser fitted with a metering valve, which is intended for disposal after use.
  • the dosage form comprises an aerosol dispenser
  • a propellant which can be a compressed gas, such as compressed air or an organic propellant, such as fluorochlorohydrocarbon.
  • the aerosol dosage forms can also take the form of a pump-atomizer.
  • Compositions suitable for buccal or sublingual administration include tablets, lozenges, and pastilles, where the active ingredient is formulated with a carrier, such as sugar, acacia, tragacanth, or gelatin and glycerine.
  • Compositions for rectal administration are conveniently in the form of suppositories containing a conventional suppository base, such as cocoa butter.
  • the compounds of the invention may be administered to an animal, e.g., a human, alone or in combination with pharmaceutically acceptable carriers, as noted above, the proportion of which is determined by the solubility and chemical nature of the compound, chosen route of administration, and standard pharmaceutical practice.
  • the dosage of the compounds of the invention, and/or compositions comprising a compound of the invention can vary depending on many factors, such as the pharmacodynamic properties of the compound; the mode of administration; the age, health, and weight of the recipient; the nature and extent of the symptoms; the frequency of the treatment, and the type of concurrent treatment, if any; and the clearance rate of the compound in the animal to be treated.
  • One of skill in the art can determine the appropriate dosage based on the above factors.
  • the compounds of the invention may be administered initially in a suitable dosage that may be adjusted as required, depending on the clinical response. In general, satisfactory results may be obtained when the compounds of the invention are administered to a human at a daily dosage of between 0.05 mg and 3000 mg (measured as the solid form). A preferred dose ranges between 0.05-500 mg/kg, more preferably between 0.5-50 mg/kg.
  • a compound of the invention can be used alone or in combination with other agents that have NOS-inhibiting activity, or in combination with other types of treatment (which may or may not inhibit NOS) to treat, prevent, and/or reduce the risk of stroke, neuropathic or migraine pain, or other disorders that benefit from NOS inhibition.
  • the dosages of one or more of the therapeutic compounds may be reduced from standard dosages when administered alone. In this case, dosages of the compounds when combined should provide a therapeutic effect.
  • a compound of the invention can also be used in diagnostic assays, screening assays, and as a research tool.
  • diagnostic assays a compound of the invention may be useful in identifying or detecting NOS activity.
  • the compound may be radiolabeled (as described elsewhere herein) and contacted with a population of cells of an organism. The presence of the radiolabel on the cells may indicate NOS activity.
  • a compound of the invention may be used to identify other compounds that inhibit NOS, for example, as first generation drugs.
  • the compounds of the invention may be used in enzyme assays and assays to study the localization of NOS activity. Such information may be useful, for example, for diagnosing or monitoring disease states or progression.
  • a compound of the invention may also be radiolabeled.
  • the compounds of the present invention have been found to exhibit selective inhibition of the neuronal isoform of ⁇ OS (n ⁇ OS).
  • Compounds may be examined for their efficacy in preferentially inhibiting n ⁇ OS over i ⁇ OS and/or e ⁇ OS by a person skilled in the art, for example, by using the methods described in Examples 19a and 19b and herein below.
  • the following non- limiting examples are illustrative of the present invention:
  • 6-amino-l-(2-(piperidin-l-yl)ethyl)-3,4-dihydroquinolin-2(lH)-one A solution of 6-nitro-l-(2-(piperidin-l-yl)ethyl)-3,4-dmydroquinolin-2(lH)-one (500mg, 1.65 mmol) in 10 mL methanol was added to Raney Nickel (slurry in H 2 O, 50 mg) in a round bottom flask. The suspension was heated at reflux for 10 minutes then filtered through a pad of celite. The celite pad was washed with 10 mL methanol.
  • N-(l-(2-(dimethylamino)ethyl)-2-oxo-l,2,3,4-tetrahydroquinolin-6-yl)thiophene- 2-carboximidamide A solution of 6-amino-l-(2-(dimethylamino)ethyl)-3,4-dihydroquinolin-2(lH)-one (0.280 g, 1.20 mmol) in absolute ethanol (5 mL) was treated with methyl thiophene-2- carbimidothioate hydroiodide (0.684 g, 2.40 mmol) at room temperature and the resulting mixture was stirred overnight (18 h).
  • This solution was transferred using a cannulating needle to a solution of l-chloro-3-iodopropane (2.52 mL, 23.47 mmol) in DMF (20 mL). The reaction was stirred at room temperature for 5 hours. The reaction was quenched with brine (25 mL), transferred to a separatory funnel and partitioned with ethyl acetate (30 mL). The aqueous was extracted twice more with ethyl acetate (2 x 20 mL). The combined organic layers were washed with brine, dried with sodium sulphate, decanted and concentrated to afford a yellow solid.
  • N-(l-(3-morpholinopropyl)-2-oxo-l,2,3,4-tetrahydroquinolin-6-yl)thiophene-2- carboximidamide A solution of 6-amino-l-(3-morpholinopropyl)-3,4-dihydroquinolin-2(lH)-one (0.055 g, 0.190 mmol) in dry ethanol (5 mL) was treated with methyl thiophene-2- carbimidothioate hydroiodide (0.108 g, 0.379 mmol) at room temperature and the resulting mixture was stirred for 64 hours.
  • N-(3-(3-(dimethylamino)propyl)-2-oxo-l,2,3,4-tetrahydroquinolin-6- yl)thiophene-2-carboximidamide dihydrochloride A solution of N-(3-(3-(dimethylamino)propyl)-l-(4-methoxybenzyl)-2-oxo- 1,2,3,4- tetrahydroquinolin-6-yl)thiophene-2-carboximidamide (50 mg, 0.1 mmol) and anisole (23 ⁇ L, 0.2 mmol) in 7.5 mL trifluoroacetic acid was stirred at room temperature for 2 hours then heated at 60 0 C for 22 hours.
  • the resulting dark brown suspension was heated at 100 0 C for 2 hours.
  • the mixture was cooled to room temperature and treated with 7 ml of a IN HCl solution then stirred at room temperature for 15 minutes.
  • the mixture was partitioned between CH 2 Cl 2 (100 mL) and IN NaOH (20 mL).
  • the organic layer was separated and the aqueous layer was extracted once more with 100 mL CH 2 Cl 2 .
  • the combined organic fractions were rinsed with brine, dried over Na 2 SO 4 , treated with -500 mg activated charcoal, filtered and concentrated to give a dark brown residue.
  • Example 19a nNOS (human), eNOS (human) and iNOS (human) Enzyme Assay
  • iNOS may be produced in Baculovirus- infected Sf9 cells (ALEXIS).
  • ALEXIS Baculovirus- infected Sf9 cells
  • NO synthase activity is determined by measuring the conversion of [ 3 H]L-arginine to [ 3 H]L-citrulline.
  • a 15 ⁇ L solution of a test substance is added to the enzyme assay solution, followed by a pre-incubation time of 15 min at RT.
  • the reaction is initiated by addition of 20 ⁇ L L-arginine containing 0.25 ⁇ Ci of [ 3 H] arginine/mL and 24 ⁇ M L-arginine.
  • the total volume of the reaction mixture is 150 ⁇ L in every well.
  • the reactions are carried out at 37°C for 45 min.
  • [ 3 H]L-citrulline is separated by DOWEX (ion- exchange resin DOWEX 50 W X 8-400, SIGMA) and the DOWEX is removed by spinning at 12,000 g for 10 min in the centrifuge. An 70 ⁇ L aliquot of the supernatant is added to 100 ⁇ L of scintillation fluid and the samples are counted in a liquid scintillation counter (1450 Microbeta Jet, Wallac). Specific NOS activity is reported as the difference between the activity recovered from the test solution and that observed in a control sample containing 240 mM of the inhibitor L-NMMA. All assays are performed at least in duplicate. Standard deviations are 10% or less. These results would show the selectivity of the compounds of the invention for nNOS inhibition. Results for exemplary compounds of the invention are also shown in Table 3 and Table 4.
  • Nitric oxide synthase neurovascular, human recombinant
  • Nitric oxide synthase endothelial, human recombinant
  • eNOS III Cat. No. ALX-201- 070, Axxora LLC
  • HPES Ethylene diamine tetra acetic acid
  • Liquid Scintillation Fluid Cat. No.6012239, Ultima Gold, Perkin-Elmer Life and
  • Vortex Mixer Mini Vortex mixer, IKA
  • Test compounds at a concentration of 6 mM are prepared from the 2 to 5 mg powder.
  • the primary stock solutions of each test compound are prepared freshly in distilled water on the day of study to obtain a final concentration of 6 mM.
  • 12 test compound concentrations are prepared as 3-fold serial dilutions.
  • Concentration range of test compound utilized for nNOS are 0.001 to 300 ⁇ M and for eNOS are 0.003 to 1000 ⁇ M.
  • the vehicle of the test compound or inhibitor is used as blank control.
  • For nonspecific activity 100 ⁇ M L-NMMA is used.
  • the IC 50 concentration of L-NAME run in parallel as controls.
  • Data is analyzed using a Sigmoidal dose-response (variable slope) curve to determine the IC 50 value of the test compound.
  • Y Bottom + (Top - Bottom)/ (1 + 10 ⁇ ((LOgIC 50 - X)*Hill Slope))
  • X is the logarithm of test compound or inhibitor concentration
  • Y is the amount of L-citrulline formation (pmol)
  • Bottom refers to the lowest Y value and Top refers to the highest Y value.
  • the slope factor also called Hill slope
  • Hill slope describes the steepness of a curve.
  • a standard competitive binding curve that follows the law of mass action has a slope of - 1.0. If the slope is shallower, the slope factor will be a negative fraction, e.g., -0.85 or -0.60.
  • Example 19b nNOS (rat), eNOS (rat) and iNOS (mouse) Enzyme Assay
  • Recombinant rat or mouse NO synthases (iNOS, eNOS, nNOS) were expressed in Sf9 cells (Sigma), 36 mg prot/ml (Bradford). Solution in 5OmM Hepes, pH 7.4 with 10% glycerol.
  • Inhibition of NOS was determined by measuring the formation of L- [3H] citrulline from L- [3H] arginine. Enzyme assays were performed in the presence of 0,25 ⁇ Ci [3H] arginine/ml, 120 ⁇ M NADPH, 1 ⁇ M FAD and FMN, 10 ⁇ M BH4, 10OnM CaM, 100 mM Hepes, 2,4 mM CaC12, 24 ⁇ M L-arginine, ImM EDTA, ImM DTT. Stop buffer: 10OmM Hepes, pH 5.5, 3mM EDTA, 3mM EGTA.
  • the efficacy of the compounds of the invention for the treatment of neuropathic pain was assessed using standard animal models predictive of anti- hyperalgesic and anti-allodynic activity induced by a variety of methods, each described in more detail below.
  • compounds of the invention are useful for treating CNS diseases.
  • a compound of the invention should be CNS penetrant.
  • a preferred example of a compound of the invention would be Example 11 over less CNS penetrant compounds such as Example 3.
  • Two compounds delivered at the same dose in the Chung animal model of neuropathic pain (a CNS disorder) differ even though the nNOS values are similar. While other factors are likely involved, activity correlates with lipophilicity.
  • (+)-Isomer (24) 2 nd eluting isomer.
  • the free base was converted to the dihydrochloride salt by standard methods.
  • tert-Butyl 3-(6-bromo-3,4-dihydroquinolin-l(2H)-yl)pyrrolidine-l-carboxylate A solution of tert-butyl 3-(3,4-dihydroquinolin-l(2H)-yl)pyrrolidine-l-carboxylate (1.86 g, 6.15 mmol) in 15 mL of DMF was cooled to 0 0 C then treated dropwise with NBS (1.09 g, 6.15 mmol) in 15 mL DMF. The reaction was stirred at 0 0 C for 1.5 hours then treated with 100 mL H 2 O. The suspension was extracted with 2 x 150 mL EtOAc.
  • N-(l-(2-(Dimethylamino)ethyl)-l,2,3,4-tetrahydroquinolin-7-yl)thiophene-2- carboximidamide (36) A solution of compound 5 (0.55 g, 2.507 mmol) in dry ethanol (10 mL) was treated with compound 6 (1.43 g, 5.015 mmol) at room temperature and the resulting mixture was stirred for over night (16 h). The reaction was diluted with sat. NaHCO 3 solution (50 mL) and product was extracted into CH 2 Cl 2 (2 x 25 mL). The combined organic layer was washed with brine (15 mL) and dried (Na 2 SO 4 ).
  • 6-Bromo-l-(l-methylpyrrolidin-3-yl)-l,2,3,4-tetrahydroquinoline 200 mg, 0.71 mmol
  • anhydrous methanol was treated with formaldehyde (37% aqueous solution, 79 ⁇ L, 1.07 mmol) followed by acetic acid (100 ⁇ L, 1.78 mmol).
  • the solution was treated with sodium cyanoborohydride (67 mg, 1.07 mmol). The suspension was stirred at room temperature for 3 hours.
  • N-(l-(l-Methylpyrrolidin-3-yl)-l,2,3,4-tetrahydroquinolin-6-yl)thiophene-2- carboximidamide A solution of 1 -( 1 -methylpyrrolidin-3-yl)- 1 ,2,3,4-tetrahydroquinolin-6-amine (80 mg, 0.35 mmol) in 8 mL ethanol was treated with methyl thiophene-2-carbimidothioate hydroiodide (197 mg, 0.69 mmol) and stirred overnight at room temperature. The mixture was diluted with CH 2 Cl 2 (10 mL) and argon was bubbled through the solution for 20 minutes.
  • Phenyl 2-(7-amino-2,3,4,5-tetrahydro-lH-benzo[b]azepin-l- yl)ethyl(methyl)carbamate A suspension of phenyl methyl(2-(7-nitro-2,3,4,5-tetrahydro- 1 H-benzo[b]azepin- 1 - yl)ethyl)carbamate (450 mg, 1.22 mmol) and palladium on activated carbon (10%, 65 mg, 0.06 mmol) in a 1 : 1 mixture of THF/EtOH (20 mL) was stirred under a balloon of hydrogen overnight. The suspension was filtered through a pad of celite.
  • 6-Amino-l-(2-(dimethylamino)ethyl)-8-fluoro-3,4-dihydroquinolin-2(lH)-one (312 mg, 1.24 mmol) was stirred to dissolve in ethanol (15 mL). To this solution was added methyl thiophene-2-carbimidothioate hydroiodide (708 mg, 2.48 mmol). The resulting suspension was stirred for 2 days at room temperature. When TLC analysis showed that the reaction was finished, the mixture was diluted with water and aqueous sodium carbonate, then extracted with dichloromethane (3x).
  • N-(2-Oxo-3-(pyrrolidin-3-yl)indolin-5-yl)thiophene-2-carboximidamide (43) A solution of compound 5 (0.21 g, 0.492 mmol) in methanol (10 mL) was treatred with 1 N HCl solution (10 mL) and the resulting solution was refluxed for 30 min. The reaction was brought to room temperature and solvent was evaporated. The crude was dissolved into water (10 mL), filtered and washed with water (2 x 5 mL). The combined water layer was evaporated to obtain compound 43 (0.173 g, 88%) as a solid.
  • Phenyl 2-(7-amino-3,4-dihydroquinolin-l(2H)-yl)ethyl(methyl)carbamate (3) A solution of compound 2 (1.85 g, 5.205 mmol) in dry ethanol (30 mL) was treated with Pd-C (-0.2 g) and purged with hydrogen gas. The reaction was stirred at room temperature under hydrogen (balloon pressure) for 3 h. The reaction was filtered through celite bed and washed with methanol (3 x 20 mL). The combined organic layer was evaporated to obtain crude compound 3 (1.67 g, 99%) as a syrup. ESI-MS (m/z, %): 326 (MH + , 100).
  • the reaction mixture was cooled to room temperature and treated with 1 N HCl (20 mL) and stirred for 15 min, then basified with 1 N NaOH (50 mL).
  • the product was extracted into CH 2 Cl 2 (2 x 50 mL) and the combined organic fractions were washed with brine (25 mL), dried (MgSO 4 ), filtered and concentrated to give a dark brown residue.
  • the crude product was purified by column chromatography (2 N NH 3 in MeOH: CH 2 Cl 2 , 5: 95) to give compound 6 (531 mg, 94%) as a syrup.
  • the resulting dark brown mixture was heated to 100 0 C and stirred for 3 hours in a sealed tube.
  • the reaction mixture was cooled to room temperature and treated with TBAF (5 mL, 1 M solution in THF) and stirred for 20 min.
  • the reaction was diluted with water (25 mL) and product was extracted into ether (3 x 50 mL) and the combined organic fractions were dried (Na 2 SO 4 ) and concentrated to give a dark brown residue.
  • the crude product was purified by column chromatography on silica gel (2 N NH 3 in methanol: CH 2 Cl 2 , 2: 98) to obtain compound 2 (0.96 g, quantitative) as a foam. .
  • tert-Butyl 3-(5-bromoindolin-l-yl)pyrrolidine-l-carboxylate (3) A solution of compound 2 (4.15 g, 14.390 mmol) in dry DMF (30 mL) was treated with NBS (2.56 g, 14.390 mmol) in dry DMF (20 mL) at O 0 C and resulting solution was stirred at same temperature for 3 h. The reaction was diluted with water (150 mL) and product was extracted into ethyl acetate (3 x 30 mL). The combined ethyl acetate layer was washed with water (2 x 25 mL), brine (25 mL) and dried (Na 2 SO 4 ).
  • N-(l-(PyrrolidinO-yl)indolin-5-yl)thiophene-2-carboximidamide (49) A solution of compound 6 (0.35 g, 0.848 mmol) in methanol (10 mL) was treated with 1 N HCl (10 mL) and the resulting mixture was refluxed for 30 minutes. The reaction was brought to room temperature and solvent was evaporated. The crude product was dissolved into water (10 mL), filtered and washed. Water was evaporated to obtain compound 49 (0.3 g, 92%) as a solid.
  • reaction mixture was cooled to room temperature and treated with 1 N HCl solution (5 mL) and stirred for 20 min.
  • the reaction was basified with 2 N NaOH solution (25 mL) and product was extracted into CH 2 Cl 2 (3 x 20 mL) and the combined organic fractions were washed with brine (20 mL) and dried (Na 2 SO 4 ).
  • the solvent was evaporated and crude product was purified by column chromatography on silica gel (2 N NH 3 in MeOH: CH 2 Cl 2 , 5: 95) to obtain compound 4 (0.44 g, 89%) as a syrup.
  • the reaction vial was sealed and dark mixture was heated at 90 °C for 2 hours.
  • the mixture was transferred to a 125 mL flask, cooled to 0 0 C and treated with 10 mL of a 2N HCl solution. After stirring for 10 minutes, the mixture was basified with IN NaOH and extracted with 2 x 75 mL CH 2 Cl 2 . After extraction, the combined organic layer was dried over Na 2 SO ⁇ filtered and concentrated to give a dark residue. This residue was subjected to flash chromatography on silica gel using 2.5% methanol / 97.5% CH 2 Cl 2 then 5% 2M NH 3 in methanol / 95% CH 2 Cl 2 to give a dark brown residue (351 mg, 89%).

Landscapes

  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Pain & Pain Management (AREA)
  • Psychiatry (AREA)
  • Psychology (AREA)
  • Addiction (AREA)
  • Rheumatology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Hospice & Palliative Care (AREA)
  • Cardiology (AREA)
  • Immunology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

La présente invention porte sur des quinolones, des tétraydroquinoléines et des composés apparentés qui inhibent l'oxyde nitrique synthase (NOS), en particulier sur celles et ceux qui inhibent de façon sélective l'oxyde nitrique synthase neuronal (nNOS) de préférence à d'autres isoformes de NOS. Les inhibiteurs de NOS de l'invention, individuellement ou en combinaison avec d'autres agents pharmaceutiquement actifs, peuvent être utilisés pour traiter ou prévenir divers états médicaux.
PCT/CA2008/000569 2007-03-23 2008-03-25 Quinolones et tétrahydroquinoléines et composés apparentés ayant une activité inhibitrice de nos WO2008116308A1 (fr)

Priority Applications (9)

Application Number Priority Date Filing Date Title
JP2010500037A JP2010521527A (ja) 2007-03-23 2008-03-25 キノロンおよびテトラヒドロキノリンならびにnos阻害活性を有する関連化合物
MX2009010193A MX2009010193A (es) 2007-03-23 2008-03-25 Quinolona y tetrahidroquinolina y compuestos relacionados que tienen actividad inhibidora de nos.
CA002681771A CA2681771A1 (fr) 2007-03-23 2008-03-25 Quinolones et tetrahydroquinoleines et composes apparentes ayant une activite inhibitrice de nos
CN200880017025A CN101679397A (zh) 2007-03-23 2008-03-25 具有nos抑制活性的喹诺酮、四氢喹啉及其相关化合物
EP08748081A EP2139886A4 (fr) 2007-03-23 2008-03-25 Quinolones et tétrahydroquinoléines et composés apparentés ayant une activité inhibitrice de nos
AU2008232269A AU2008232269A1 (en) 2007-03-23 2008-03-25 Quinolone and tetrahydroquinoline and related compounds having NOS inhibitory activity
NZ580618A NZ580618A (en) 2007-03-23 2008-03-25 Quinolone and tetrahydroquinoline and related compounds having nos inhibitory activity
IL201142A IL201142A (en) 2007-03-23 2009-09-23 Quinolone and tetrahydroquinoline and related compounds have nos inhibitory activity
ZA2009/07458A ZA200907458B (en) 2007-03-23 2009-10-23 Quinolone and tetrahydroquinoline and related compounds having nos inhibitory activity

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US89682907P 2007-03-23 2007-03-23
US60/896,829 2007-03-23

Publications (1)

Publication Number Publication Date
WO2008116308A1 true WO2008116308A1 (fr) 2008-10-02

Family

ID=39775375

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2008/000569 WO2008116308A1 (fr) 2007-03-23 2008-03-25 Quinolones et tétrahydroquinoléines et composés apparentés ayant une activité inhibitrice de nos

Country Status (14)

Country Link
US (1) US8173813B2 (fr)
EP (1) EP2139886A4 (fr)
JP (1) JP2010521527A (fr)
CN (1) CN101679397A (fr)
AR (1) AR065845A1 (fr)
AU (1) AU2008232269A1 (fr)
CA (1) CA2681771A1 (fr)
CL (1) CL2008000838A1 (fr)
IL (1) IL201142A (fr)
MX (1) MX2009010193A (fr)
NZ (1) NZ580618A (fr)
TW (1) TWI417099B (fr)
WO (1) WO2008116308A1 (fr)
ZA (1) ZA200907458B (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8198268B2 (en) 2008-10-31 2012-06-12 Janssen Biotech, Inc. Tianeptine sulfate salt forms and methods of making and using the same
US10092591B2 (en) 2014-02-27 2018-10-09 University Of Alaska Fairbanks Methods and compositions for the treatment of ischemic injury to tissue using therapeutic hypothermia

Families Citing this family (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NZ586082A (en) * 2007-11-16 2013-03-28 Neuraxon Inc Indole compounds and methods for treating visceral pain
WO2009064505A1 (fr) * 2007-11-16 2009-05-22 The Arizona Board Of Regents On Behalf Of The University Of Arizona Procédés de traitement de la douleur viscérale
US20110172223A1 (en) * 2008-09-29 2011-07-14 Chakravarty Prasun K Substituted Aryl Sulfone Derivatives as Calcium Channel Blockers
WO2010088408A2 (fr) * 2009-01-28 2010-08-05 Emory University Antagonistes du récepteur nmda, sélectifs pour des sous-unités, et destinés au traitement d'états neurologiques
FR2984318B1 (fr) 2011-12-16 2014-06-27 Oreal Coupleur de structure 7 amino-1,2,3,4-tetrahydroquinoleines cationiques, composition tinctoriale en comprenant, procedes et utilisations
FR2984323B1 (fr) 2011-12-16 2019-08-30 L'oreal Coupleur de structure 7 amino-1,2,3,4-tetrahydroquinoleines, composition tinctoriale en comprenant, procedes et utilisations
CA2896871A1 (fr) * 2012-12-31 2014-07-03 Kerry L. Spear Composes heterocycliques et leurs procedes d'utilisation
EP2986587B1 (fr) * 2013-04-17 2023-11-22 Sharon Anavi-Goffer Ligands du récepteur cb2 pour le traitement de troubles psychiatriques
WO2014208586A1 (fr) * 2013-06-26 2014-12-31 第一三共株式会社 Procédé de production d'un composé hétérocyclique tricyclique
CA2919892C (fr) 2013-08-12 2019-06-18 Pharmaceutical Manufacturing Research Services, Inc. Comprime extrude anti-abus a liberation immediate
US10172797B2 (en) 2013-12-17 2019-01-08 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
US9492444B2 (en) 2013-12-17 2016-11-15 Pharmaceutical Manufacturing Research Services, Inc. Extruded extended release abuse deterrent pill
AU2015290098B2 (en) 2014-07-17 2018-11-01 Pharmaceutical Manufacturing Research Services, Inc. Immediate release abuse deterrent liquid fill dosage form
CA2964628A1 (fr) 2014-10-20 2016-04-28 Pharmaceutical Manufacturing Research Services, Inc. Forme galenique anti-abus de remplissage de liquide a liberation prolongee
CN109206385B (zh) * 2017-07-03 2022-04-22 扬子江药业集团有限公司 一种地佐辛杂质a及其同系物的制备方法
FR3072286B1 (fr) 2017-10-13 2022-08-12 Oreal 7-amino-1,2,3,4-tetrahydroquinoleines particuliers, procede et composition
CN108853509A (zh) * 2018-05-03 2018-11-23 浙江大学 抑郁症的治疗和药物组合物
CN109053712A (zh) * 2018-09-19 2018-12-21 南京大学连云港高新技术研究院 一种2-氨基噻吩-3-羧酸甲酯衍生物c15h22n2o5s的合成方法
WO2020117899A1 (fr) 2018-12-05 2020-06-11 Northwestern University Inhibiteurs de l'oxyde synthase neuronal pour l'immunothérapie
US20230057844A1 (en) * 2019-12-20 2023-02-23 The Jackson Laboratory Molecular targets for addiction
CN111650307A (zh) * 2020-07-13 2020-09-11 重庆医药高等专科学校 一种同时对酒中解热镇痛类药品非法添加物定性定量检测的方法及应用
WO2022056736A1 (fr) * 2020-09-16 2022-03-24 中国科学院深圳先进技术研究院 Application de lévofloxacine ou de sels pharmaceutiquement acceptables de cette dernière dans la préparation de médicaments contre les lésions cérébrales d'ischémie-reperfusion ou de produits de soins de santé
CN115073370A (zh) * 2021-03-10 2022-09-20 成都硕德药业有限公司 新型烷基氨类化合物或盐、异构体、其制备方法及用途
WO2023177823A1 (fr) * 2022-03-17 2023-09-21 Vasoceuticals, Inc. Utilisation combinée de composés individuels pour le traitement de troubles de la douleur chronique

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5373571A (en) * 1976-12-10 1978-06-30 Otsuka Pharmaceut Co Ltd Carbostyril derivatives
CA1069753A (fr) * 1975-12-22 1980-01-15 Kekhusroo R. Bharucha Regulation de la formation de la nitrosamine dans les viandes preparees, a l'aide de composes de type 1,2,3,4-tetrahydroquinoline
CA1081222A (fr) * 1976-03-31 1980-07-08 Roussel-Uclaf Procede de preparation de nouvelles benzazepines et leurs sels
CA1169428A (fr) * 1980-08-12 1984-06-19 David E. Bays Derives d'indole
CA1253497A (fr) * 1983-12-22 1989-05-02 David A. Roberts Agents inotrope a base de quinolone heterocyclique substituee
EP0322746A1 (fr) * 1987-12-30 1989-07-05 Orion Corporation Limited Composés hétérocycliques
WO1998023610A1 (fr) * 1996-11-25 1998-06-04 The Procter & Gamble Company Composes 2-imidazolinylaminoindole convenant comme agonistes de l'adrenorecepteur alpha-2
US5854234A (en) * 1993-10-21 1998-12-29 G. D. Searle & Co. Amidino dervatives useful as nitric oxide synthase inhibitors
WO2003045313A2 (fr) 2001-11-27 2003-06-05 Merck & Co. Inc. Composés de 2-aminoquinoline
WO2003076405A1 (fr) * 2002-03-14 2003-09-18 Bayer Healthcare Ag Aroylpyridinones monocycliques tenant lieu d'agents anti-inflammatoires
US20060258721A1 (en) 2005-04-13 2006-11-16 Shawn Maddaford Substituted indole compounds having NOS inhibitory activity
CA2615007A1 (fr) * 2005-07-13 2007-01-18 F. Hoffmann-La Roche Ag Derives de benzimidazole tels que 5-ht6, 5-ht24

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS57192389A (en) 1981-05-20 1982-11-26 Takeda Chem Ind Ltd Novel maytansinoid
US4963677A (en) * 1987-12-30 1990-10-16 Orion Corporation Ltd. Heterocyclic lactam compounds
US5847006A (en) * 1991-02-08 1998-12-08 Cambridge Neuroscience, Inc. Therapeutic guanidines
CA2182302A1 (fr) * 1994-02-03 1995-08-10 Stanley M. Goldin Guanidines therapeutiques
MX9605366A (es) * 1994-05-07 1997-12-31 Astra Ab Derivados de amidina biciclicos como inhibidores de la oxido nitrico sintetasa.
CN1104422C (zh) * 1996-11-25 2003-04-02 普罗克特和甘保尔公司 用作α-2肾上腺素能受体兴奋剂的胍基杂环化合物
JPH10265450A (ja) * 1997-03-25 1998-10-06 Mitsui Chem Inc 一酸化窒素合成酵素阻害作用を有する新規なアミジン誘導体
US6166030A (en) * 1997-05-05 2000-12-26 Astra Aktiebolag Compounds
FR2783519B1 (fr) * 1998-09-23 2003-01-24 Sod Conseils Rech Applic Nouveaux derives d'amidines, leur preparation, leur application a titre de medicaments et les compositions pharmaceutiques les contenant
JP2004536079A (ja) * 2001-05-16 2004-12-02 アンテックスファーマ・インコーポレーテッド 置換1−ベンズアゼピンおよびその誘導体
CA2370751A1 (fr) * 2002-02-05 2003-08-05 Garth B.D. Shoemaker Methode de rendu rapide et precise de distorsions en technologie d'ecran pliable faisant appel a des couvertures en texels precalculees
US20050171195A1 (en) * 2003-11-06 2005-08-04 Carroll William A. P2X7 antagonists for treating neuropathic pain
CA2607219A1 (fr) * 2005-05-18 2007-02-15 Neuraxon, Inc. Composes de benzimidazole substitues a action a la fois inhibitrice de nos et agoniste opioide mu

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA1069753A (fr) * 1975-12-22 1980-01-15 Kekhusroo R. Bharucha Regulation de la formation de la nitrosamine dans les viandes preparees, a l'aide de composes de type 1,2,3,4-tetrahydroquinoline
CA1081222A (fr) * 1976-03-31 1980-07-08 Roussel-Uclaf Procede de preparation de nouvelles benzazepines et leurs sels
JPS5373571A (en) * 1976-12-10 1978-06-30 Otsuka Pharmaceut Co Ltd Carbostyril derivatives
CA1169428A (fr) * 1980-08-12 1984-06-19 David E. Bays Derives d'indole
CA1253497A (fr) * 1983-12-22 1989-05-02 David A. Roberts Agents inotrope a base de quinolone heterocyclique substituee
EP0322746A1 (fr) * 1987-12-30 1989-07-05 Orion Corporation Limited Composés hétérocycliques
US5854234A (en) * 1993-10-21 1998-12-29 G. D. Searle & Co. Amidino dervatives useful as nitric oxide synthase inhibitors
WO1998023610A1 (fr) * 1996-11-25 1998-06-04 The Procter & Gamble Company Composes 2-imidazolinylaminoindole convenant comme agonistes de l'adrenorecepteur alpha-2
WO2003045313A2 (fr) 2001-11-27 2003-06-05 Merck & Co. Inc. Composés de 2-aminoquinoline
WO2003076405A1 (fr) * 2002-03-14 2003-09-18 Bayer Healthcare Ag Aroylpyridinones monocycliques tenant lieu d'agents anti-inflammatoires
US20060258721A1 (en) 2005-04-13 2006-11-16 Shawn Maddaford Substituted indole compounds having NOS inhibitory activity
CA2615007A1 (fr) * 2005-07-13 2007-01-18 F. Hoffmann-La Roche Ag Derives de benzimidazole tels que 5-ht6, 5-ht24

Non-Patent Citations (13)

* Cited by examiner, † Cited by third party
Title
ABDEL-MAJID ET AL., J. ORG. CHEM., vol. 61, 1996, pages 3849 - 3862
AM. J. PHYSIOL., vol. 268, 1995, pages R286
ARROWSMITH ET AL., STROKE, vol. 29, 1998, pages 2357 - 2362
BELETSKAYA; CHEPRAKOV, CHEM. REV., vol. 100, pages 3009 - 3066
DAWSON ET AL., PROC. NATL. ACAD. SCI., vol. 90, no. 8, 1993, pages 3256 - 3259
HICKS ET AL., EUR. J. PHARMACOL., vol. 381, 1999, pages 113 - 119
J. MED. CHEM., vol. 47, 2004, pages 2973 - 2976
PARMENTIER ET AL., BR. J. PHARMACOL., vol. 127, 1999, pages 546
RICHARDSON A.: "The Chemistry of 7-Aminoindoline and Certain Pyrrolo- and Pyrido[1,2,3-de]quinoxalines", J. ORG. CHEM., vol. 30, no. 8, 1965, pages 2589 - 2593, XP002512077 *
ROCH ET AL., N ENG. J. MED., vol. 335, 1996, pages 1857 - 1864
See also references of EP2139886A4
SHAW ET AL., Q. J. MED., vol. 58, 1986, pages 59 - 68
WINKELMANN ET AL.: "Tuberkulostatisch Wirksame N,N'-diarylthioharnstoffe", ARZNEIMITTEL-FORSCHUNG, vol. 19, no. 4, 1969, pages 543 - 558, XP008120126 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8198268B2 (en) 2008-10-31 2012-06-12 Janssen Biotech, Inc. Tianeptine sulfate salt forms and methods of making and using the same
US10092591B2 (en) 2014-02-27 2018-10-09 University Of Alaska Fairbanks Methods and compositions for the treatment of ischemic injury to tissue using therapeutic hypothermia

Also Published As

Publication number Publication date
US8173813B2 (en) 2012-05-08
US20080234237A1 (en) 2008-09-25
CA2681771A1 (fr) 2008-10-02
TW200902003A (en) 2009-01-16
AR065845A1 (es) 2009-07-08
NZ580618A (en) 2012-07-27
AU2008232269A1 (en) 2008-10-02
MX2009010193A (es) 2009-12-15
EP2139886A4 (fr) 2011-03-30
IL201142A (en) 2013-12-31
TWI417099B (zh) 2013-12-01
EP2139886A1 (fr) 2010-01-06
IL201142A0 (en) 2010-05-17
JP2010521527A (ja) 2010-06-24
CL2008000838A1 (es) 2008-10-10
CN101679397A (zh) 2010-03-24
ZA200907458B (en) 2011-01-26

Similar Documents

Publication Publication Date Title
US8173813B2 (en) Quinolone and tetrahydroquinolone and related compounds having NOS inhibitory activity
CA2729246C (fr) Benzoxazines, benzothiazines et composes apparentes ayant une activite d'inhibition de nos
AU2006321284B2 (en) Substituted indole compounds having NOS inhibitory activity
US20090131503A1 (en) 3,5 - substituted indole compounds having nos and norepinephrine reuptake inhibitory activity
US7919510B2 (en) Substituted benzimidazole compounds with dual NOS inhibitory activity and mu opioid agonist activity
AU2007240078B2 (en) 1,5 and 3,6- substituted indole compounds having NOS inhibitory activity

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200880017025.5

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08748081

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2681771

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 201142

Country of ref document: IL

Ref document number: MX/A/2009/010193

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2010500037

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2008232269

Country of ref document: AU

Ref document number: 580618

Country of ref document: NZ

REEP Request for entry into the european phase

Ref document number: 2008748081

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 6796/DELNP/2009

Country of ref document: IN

Ref document number: 2008748081

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2008232269

Country of ref document: AU

Date of ref document: 20080325

Kind code of ref document: A